You are on page 1of 21

Biochemical Engineering Journal 133 (2018) 219–239

Contents lists available at ScienceDirect

Biochemical Engineering Journal


journal homepage: www.elsevier.com/locate/bej

Review

Challenges and opportunities in lactic acid bioprocess design—From


economic to production aspects
Regiane Alves de Oliveira a,∗ , Andrea Komesu b , Carlos Eduardo Vaz Rossell c ,
Rubens Maciel Filho a
a
Laboratory of Optimization, Design and Advanced Process Control, School of Chemical Engineering, University of Campinas—Unicamp, 500 Albert Einstein
Av, Campinas, 13083-852, Brazil
b
Department of Marine Sciences, Federal University of São Paulo—UNIFESP, 144 Carvalho de Mendonça St, Santos, 11070-100, Brazil
c
Interdisciplinary Center of Energy Planning (NIPE), University of Campinas - Unicamp, 330 Cora Coralina St, Campinas, 13083-896, Brazil

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

Article history: Lactic acid is an already consolidated bioproduct in the world market. It has many applications, such as:
Received 9 August 2017 the production of biodegradable polymers, substitution of plastics from oil, and new uses in medicine.
Received in revised form 28 February 2018 Besides this, new applications are being discovered every year, especially in chemical industries as a
Accepted 3 March 2018
building-block molecule. The lactic acid market is in constant growth and the fact that the final products
Available online 7 March 2018
are able to comply with environmental laws as green, renewable and biodegradable products contributes
to the tendency of continuous growth in the next few years. With all of this in mind, this paper will explore
Keywords:
the main aspects of the sector, as well as market tendencies, production chain, and innovation.
Lactic acid
Applications © 2018 Elsevier B.V. All rights reserved.
Global market
Feedstock
Lactic acid bacteria
Downstream

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220
2. New lactic acid applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 220
3. Global market and techno-economic aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
3.1. Corbion–Purac (Netherlands) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
3.2. Galactic (Belgium) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
3.3. NatureWorks LLC – Cargill (USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
3.4. Others . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
4. Lactic acid biorefineries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
5. Life-cycle assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223
6. Fermentation feedstock for lactic acid production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 225
6.1. Starchy materials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
6.2. Lignocellulosic biomass . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
6.3. Microalgae . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
6.4. Food waste . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
6.5. Glycerol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 226
6.6. Feedstocks concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
7. Lactic acid bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
7.1. Nutritional requirements for lactic acid bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228

∗ Corresponding author. Tel.: +55 19983490710.


E-mail address: raoliveira@feq.unicamp.br (R. Alves de Oliveira).

https://doi.org/10.1016/j.bej.2018.03.003
1369-703X/© 2018 Elsevier B.V. All rights reserved.
220 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

7.2. Choosing micro-organisms – desirable characteristics of Lactobacillus sp. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228


7.3. Metabolic pathways for lactic acid production by Lactobacillus sp. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
7.4. Main metabolic characteristics of Lactobacillus sp. under stress conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 230
7.5. Genetically engineering lactic acid bacteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232
8. Lactic acid separation and purification – downstream processes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233
9. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
WEB references . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239

1. Introduction ogy, chemistry, genomics and molecular biology to work together,


in order to reach a sustainable and economically viable production
The lactic acid molecule contains a chiral carbon and may be chain of lactic acid for supplying the market.
found in two different isomeric forms: L and D. The proportion of
each isomer confers different physical properties to the final prod- 2. New lactic acid applications
uct. It makes enantiomeric purity a crucial factor for lactic acid
industrial production, as in the manufacture of biodegradable plas- Lactic acid has a wide range of industrial applications as shown
tics and polymers as poly(lactic acid) (PLA). The D-lactic acid isomer in Fig. 1. It is a high value-added product that has been gain-
in high doses is considered harmful to humans and can cause aci- ing market share with each year [17–19]. Recently, it has been
dosis or de-calcification [1,2]. Therefore, the L-lactic acid isomer is highlighted in the environmental, ecological, and medical areas,
preferred by the pharmaceutical and food industries [3]. This fact especially as a building-block molecule. In addition, it has been used
corresponded to a higher demand for the product at the beginning for a long time in the food industry. When lactic acid is produced
of the 2000s [4]. Demand continues to increase due to many medical by homo-fermentation, it is possible to achieve high selectivity and
applications that make use of PLA [5]. conversion with no CO2 delivery.
It is well known that chemical production leads to racemic mix- In the food industry, lactic acid is well known because it can be
tures of DL-lactic acid [4,6–8]. This fact makes it impossible to used in various forms as an additive. It may also result from the
control the chemical and physical properties of the final product. It presence of a contaminant micro-organism in some types of food.
is also unsuitable for use in the food, pharmaceutical, and medical It is used mainly in the isomeric form L, since this is the only form
industries. that the human body is adapted to assimilate [11,13]. It is employed
Some industries require a high enantiomeric purity of lactic acid as an additive, acidulant, flavouring and emulsifier, apart from also
for specific applications. This fact also contributes to lactic acid inhibiting sporulation of bacteria in processed foods [3,20–22].
production via fermentation being more attractive instead of chem- For the pharmaceutical industry, lactic acid is used in the pro-
ical production. This is especially relevant when lactic is used as a duction of cosmetics, formulating ointments, anti-acne solutions,
monomer for polymers applied in the medical, pharmaceutical and humectants, and controlled-release drugs [1,3,20,21,23]. In this
food industries.
Lactic acid production via fermentation has several advantages
when compared to chemical syntheses, such as: low-cost sub-
strates, relatively lower temperatures, lower energy consumption,
better environmental concerns, high purity [9–11] plus ease of cre-
ating products with tailor-made characteristics.
Industrially, about 90% of all lactic acid produced worldwide is
derived from bacterial fermentation [12–15]. The most widely used
method of lactic acid production is batch fermentation. The yield
of lactic acid in this case is 90–95% w/w, according to the initial
sugar content used [5]. During lactic acid fermentation, the main
parameters to be considered are: pH, temperature, initial substrate
concentration, and nitrogen concentration. These variables directly
affect the rate and therefore the performance of the fermentation
[4,16].
The lactic acid industry has many challenges to be overcome
in the following years. In regards to the fermentation process, the
biggest challenges are: the price of the substrates, food compe-
tition, strains, inhibition due to substrate or product, and waste
generation. In addition, the process of separation and purification
is crucial for this development, since many applications require a
product with high purity, such as the medical and pharmaceuti-
cal areas. Along with these challenges come environmental issues,
with increasingly severe legislation regarding the use of solvents
and waste generation, seeking a sustainable production chain.
This paper intends to discuss the current state of lactic acid pro-
duction and the importance of the connection between distinct
areas in the development of a productive chain of lactic acid. It
shows the importance of connecting the areas of engineering, biol-
Fig. 1. Lactic acid industrial applications.
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 221

case, the L-lactic acid isomer is also the most used because of its 3. Global market and techno-economic aspects
compatibility with the human body.
In the chemical industry, lactic acid has gained ground every Lactic acid is commonly sold as an 88% solution [37]. The
year. Its versatility of applications has created a positive outlook price varies with the application (e.g., food, pharmaceuticals, and
for investments, enabling the emergence of new products based on PLA) and a range of $1.30–$2.30/kg was reported in 2011, nowa-
the molecule (Fig. 1). It has a wide range of applications, such as the days reaching around $3.0–$4.0/kg (https://www.pharmacompass.
production of organic solvents [3,20] and building-block molecules com). In general, the price of lactic acid will follow the price of
[24]. Furthermore, motivated by environmental concerns at the end commodity starch and sugar feedstocks used for fermentation [37].
of the 90s, lactic acid has emerged as a potential chemical com- The global lactic acid market required 1,220.0 kt in 2016. With
pound for the production of renewable and biodegradable plastics annual growth of 16.2%, it is expected that this demand will reach
from sugar fermentation [1,3,25–27]. The highlight of renewable 1,960.1 kt in 2025. This should represent USD 9.8 billion in the
and biodegradable plastics from lactic acid has existed since the global market. Market studies point out that the largest and fastest
early 2000s in the production of poly(lactic acid) (PLA) [28] for growth in this period will be for medicines and perfumes in the
production of food packaging and many plastic utensils, which Latin America and the Asia Pacific region. Latin America’s lactic
replaced products made from raw petroleum [1,29]. The monomer acid market is experiencing significant growth – 19.2% annually
for the production of PLA provides a wide versatility of use and – mainly because of the growth of the cosmetics industry in Brazil
favourable characteristics, such as: biodegradability, biocompati- and Argentina. As with lactic acid, PLA demand should grow, reach-
bility, elasticity, and also a well-controlled release profile of drugs ing USD 6.5 billion worldwide by 2025. The reason for this would be
[23,30]. PLA is considered one of the most promising biodegrad- the Asia Pacific PLA market growth rate of 22.4% annually, because
able plastics [28], mainly due to its high chemical resistance, which of the establishment of various food and beverage as well as cos-
is advantageous for the manufacture of fibre and film, while its metics manufacturing units, particularly in India and China (http://
heat resistance is favourable for the production of many uten- www.grandviewresearch.com).
sils, such as cups and trays [31,32]. However, its use in industry According to another source, the lactic acid market represented
when produced via the biochemical pathway is still restricted USD 1,275.00 million in 2014, and is expected to reach USD 4,129.19
due to the high cost of lactic acid production when compared million by 2022. This would represent an annual growth of 16.9%
with petrochemical compounds [33]. These expenses are always from 2015 to 2022. In terms of volume, 2014 required around 36 kt
associated with production steps such as fermentation and down- more lactic acid than 2013 and is expected to reach 1,844.56 kt by
stream. 2022, with annual growth of 12.9% (http://www.wtva.com).
Another promising field of application for lactic acid at the By 2019, the lactic acid market is expected to exceed USD 3,577.5
moment is the medical area, with use in tissue regeneration, million. The figure expected for PLA is USD 4,840.1 million. Europe
surgical sutures, fracture fixation, implants and ligaments, car- should represent the main market for PLA, while the Asia Pacific
tilage repair, meniscus, bone replacement, and oral surgeries. region should represent the fastest growing market (http://www.
These uses are already approved by the American Food and Drug bioplasticsmagazine.com).
Administration (FDA) [23,34,35]. PLA can be also used in the In fact, all the analysts in the sector have made positive predic-
form of screws, pins, clamps, and boards. The main advantages tions for both the lactic acid and PLA markets for the next years.
of PLA prostheses and implants are their high strength, lack of There is a sense of optimism from producers, especially due to
rejection, and the ability of being completely reabsorbed by the the fact that new applications have been appearing constantly in
body. It makes surgeries to remove patients’ implants unneces- the market. Only in 2017, Galactic, one of the largest producers
sary [36], which reduces expenses with medical procedures and in the world, announced five new products on its website (https://
facilitates patient recovery. For these applications, the fermen- www.lactic.com). One of the major drivers influencing the growing
tation route is very suitable. In this case, products with specific market is expected to be the increasing preference of consumers
characteristics are required, and it depends on the lactic acid iso- towards eco-friendly packaging, leading more brands to adopt
mer used. PLA and its copolymers have been in clinical use for biodegradable materials for packaging [37]. Nevertheless, high pro-
many years, and have been proven to be biocompatible and safe duction costs will prove to be a major challenge, as crude oil-based
[23]. polymers are less expensive when compared to PLA (http://www.
In summary, lactic acid has applications in the following sectors: prnewswire.com).
personal and home care, biodegradable plastics, as a building- Techno-economic evaluations of new processes have been
block for poly(lactic acid) (PLA), as a biodegradable polymer, developed for the market, and have shown generally positive
food and beverage (fermentation agent, food preservative and results for the implementation of such new processes regarding
de-contaminant, nutrition fortifier, acidulant, flavour enhancer, lactic acid production. A study regarding different applications for
acidic nature of selected food-grade organic acids), pharmaceuti- food waste concluded that a biorefinery producing plasticizer and
cals, mosquito repellent, many industrial applications, and animal lactic acid would be economically feasible, with annual net prof-
health. The newest achievements are related to applications its of around US$ 422,699, a present net value of US $ 3,028,000,
of heteropolysaccharides (HePS), immune-stimulant, antioxidant, with a payback period of 7.56 years and an internal rate of return
cryoprotectant, pre-biotic activity, and viscosifier (http://markets. of 18.98% [38]. In another case, the most suitable scenario was
businessinsider.com). the co-production of lactic acid and ethanol, also in a biorefinery
Key companies are constantly introducing new uses and prod- sense [39]. Many techno-economic evaluations are developed for
ucts in the market as “Graphene-enhanced PLA Filaments for 3D different processes and with different substrates [40–45], and for
Printing; PLA Formulation for 3D Printing Filament Providing High most of them, the most expensive income is related to substrates or
Impact Strength and Heat Resistance; PLA Resin from Second Gen- nutrients [40,44,45], which are also related to the geographic loca-
eration Feedstocks; a Unique, High-Quality grade L-Lactic Acid”. tion of the production facility. Details from some techno-economic
Also, NatureWorks claims to be developing a commercial scale evaluations from literature are shown in Table 1.
methane to lactic acid conversion (http://markets.businessinsider. A recent study found the minimum selling price for lactic acid
com). should be 0.56 US$/kg [41]. In this case, the authors used a final
222 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

Table 1
Techno-economic analysis of lactic acid production from renewable resources according to literature.

Substrate Downstream process Overall scenario Minimum sell price Reference

Lignocellulosic residues, Hemicellulose liquid fraction Reactive distillation 2nd best scenario: LA 1.30–5.00 US$/kg [42]
sugarcane bagasse and Ca(OH)2 production rates from
leaves. cellu-lignin and Ca(OH)2
Pretreatment: Steam explosion Cellu-lignin solid fraction
Ca(OH)2
Hemicellulose liquid fraction 3rd best scenario: LA
Acid tolerant bacteria production rates from
Cellu-lignin solid fraction cellu-lignin and acid tolerant
Acid tolerant bacteria bacteria
Hemicellulose liquid fraction 1st best scenario: LA
Mg(OH)2 production rates from
Cellu-lignin solid fraction cellu-lignin and Mg(OH)2
Mg(OH)2
Sugarcane juice Nanofiltration The two largest cost 3.15 US $/kg, 80% (w/w), and [40]
Uncontrolled pH components: raw material and 95% purity
yeast extract (6% and 87%
respectively)
Sweet cheese whey ultrafiltration permeate NaOH Ultrafiltration, ion exchange, Fermentation step requires the 1.25 US$/kg, 50% (w/w), 99% [44]
reverse osmosis, and vacuum highest operating cost (47%), purity, and racemic mixture
evaporation yeast extract is the major
contributor to the operating
costs (25%)
Whole-wheat flour NaOH Ultrafiltration, electrodialysis, The costs of raw material, the 0.83 US$/kg, 70% (w/w) [45]
and evaporation NaOH in the fermentation step,
and the conversion of lactate to
LA using electrodialysis were
the major production cost.
Batchwise step was
economically better than
continuous fermentation.
Biodetoxified corn stover. – Not considering C5 [41]
0.56 US$/kg, 88% (w/w)
Pretreatment: Diluted acid and enzymes Ca(OH)2 consumption. Highest
operating cost was celullase

lactic acid concentrated at 88% (w/w), produced from corn stover have dedicated themselves to its production and use in several
hydrolysate. The pretreatment used was diluted acid, followed industrial areas. Industry-leading companies are located in devel-
by an enzymatic hydrolysis. They did not consider C5 sugar con- oped countries, focusing their resources on R & D to serve the main
sumption, and the acid neutralization was made using Ca(OH)2 . consumer markets. Among them, the following stand out (sorted
Evaluating this process, the authors described cellulase as the most alphabetically):
expensive step in the entire process [41].
Regarding the downstream process, the traditional process for
lactic acid recovery and purification is still used: fermentation broth 3.1. Corbion–Purac (Netherlands)
is neutralized with calcium hydroxide, which forms calcium lactate
salt; the solution is filtered to remove microbial cells and evapo- This company is a well-known lactic acid producer on the mar-
rated to concentrate; sulphuric acid is added to hydrolyse calcium ket. It supplies different sectors, from food, chemicals, plastics
lactate, resulting in insoluble gypsum; methanol is added to ester- (made from PLA), biomedicine, and beyond (http://www.corbion.
ify the lactic acid and the final product is purified by distillation com, A). The manufacturer produces L-lactic acid and D-lactic acid
and hydrolysis [37]. Many different possibilities are reported in the with high purities to be used in pharmaceutical and PLA sectors
literature [37,46], but the majority still face problems with costs. A (http://www.corbion.com, B). The company claimed the patent for
possible substitute for the traditional process is the reactive distil- gypsum-free lactic acid production in 2014, and has also been
lation [47]. This process has been described in the literature as one working on the production of lactic acid for PLA from second-
of the most promising on an industrial scale, also presenting some generation feedstocks (http://www.corbion.com, C). The market
cost advantages [42,48]. The process consists of esterification of of Corbion is divided into: 74.2% (net sales) for bakery supplies,
lactic acid with methanol or another alcohol, followed by hydrol- 23.4% for biochemical products, and 2.4% to others. The net sales
ysis of the separated methyl lactate, resulting in pure lactic acid are distributed across the Netherlands (11.2%), Europe (7.2%), North
obtained from the bottom of the reactive distillation column [47]. America (63.3%) and others (18.3%) (https://www.euronext.com).
In the techno-economic evaluation performed by Daful et al. [42],
the authors considered the approach using ethanol in an integrated
biorefinery context with very successful results. 3.2. Galactic (Belgium)
To boost the competitiveness of the lactic acid industry with
products from oil, the cost must be kept down, in consistency Operating for more than 20 years, this company is one of the
with products from fossil sources [4,29]. Exceptions would occur in leaders in biotechnology, mainly in the food, feed, personal and
cases with more specific applications, requiring properties of lactic healthcare, and industrial markets. One of the aims of lactic acid
acid obtained by fermentation, such as products with higher added fermentation and the development of many other derivatives is to
value [49,50]. create sustainable, innovative and health-friendly solutions in the
To sum up, since the 90s lactic acid has been gaining ground field of food safety, nutrition and green chemistry (https://www.
in the market through its diverse applicability. Large companies lactic.com, A). In constant expansion, the release of new products
in 2017 is related to fish and seafood manufacture, facial lotions
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 223

and hand creams, bakery products, biossolvents, beverages, and [26], food waste [64,65], algal biomass [66], wheat bran [67], cof-
detergents (https://www.lactic.com, B). fee pulp [68], and others. Details of some processes are shown in
Table 2.
3.3. NatureWorks LLC – Cargill (USA) A fundamental aspect regarding these biorefineries is connected
to the fact that the processes developed and the goals of the prod-
Considered one of the world leaders on lactic acid supply, using uct are extremely dependent on the location of the site [55]. This is
the brad IngeoTM , the facility in Nebraska, USA, has a nameplate one of the drivers of the way a biorefinery will operate to achieve
capacity of 140,000 metric tons of IngeoTM biopolymer for rigid the economic, social and environmental goals. In addition, one of
and flexible packaging solutions, food service ware, health and per- the main challenges facing the growing bioeconomy is to develop
sonal care, durable products in home, appliance, and electronic efficient and cost-effective green technologies for sustainable pro-
categories, and 3D printing filament (http://www.natureworksllc. duction of commodity items from biomass, such as lactic acid.
com). The company recently announced Thailand as a preferred Some examples proposing the integration of lactic acid biore-
location for a second manufacturing plant, although the main R & finery with other products are shown below:
D facility will remain in the USA (https://www.industryintel.com).
• Brewer spent grains on a biorefinery concept to produce lactic
3.4. Others
acid, xylitol, activated carbon, and phenolic acids. The authors
performed full mass integration on water and full energy inte-
ADM; B&G; BASF; Cellulac; Danimer Scientific; Direvo Indus-
gration to achieve a configuration with the best economic and
trial Biotechnology; Dow; ESUN; Futerro; Henan Jindan Lactic
environmental performance [43];
Acid Technology; Hisun Biomaterials; Jiuding Biological Engi- • Food waste enhancement using fungal hydrolysis, and microalgae
neering; Myriant; Musashino Chemical; Piaoan; Nantong Jiuding
cultivation. The authors proposed producing plasticizer and lactic
Biological Engineering; Pyramid Bioplastics; Shenzhen BrightChina
acid which were found to be economically feasible [38];
Industrial; Synbra Technology; Teijin; ThyssenKrupp Industrial • Sugarcane lignocellulose biorefineries annexed to a typical sugar
Solutions; Tongjieliang; Uhde Inventa-Fischer; Yangtzelabre; Zhe-
mill to co-produce ethanol, lactic acid, and/or electricity. Lactic
jiang Hisun Chemical are some of the companies with activities in
acid co-production with sugar was the most economically attrac-
lactic acid production (https://firstnewshawk.com) [37].
tive scenario. However, ethanol production from xylose and lactic
Musashino Chemical Laboratory, Ltd. in Japan is a producer of
acid from glucose was found to be the most desirable biorefinery
synthetic lactic acid, with a total production of 7000 metric tons
scenario, by balancing financial and environmental considera-
per year, or about 2% of global lactic acid production [37].
tions [69];
• Integrated biorefinery system combining the production of
4. Lactic acid biorefineries
bioethanol from winter wheat-straw and lactic acid from alfalfa.
In this case, the authors performed the exchange of useful energy
In the last years, biochemicals have rapidly gained market share,
necessary for biomass processing in the production chain, con-
competing with traditional oil resources, since petrochemicals are
cluding that this approach had a better performance in the LCA
finite [51] and sometimes suffer price fluctuation that is not con- impact categories than both standalone systems [70].
ducive to successful economic planning.
Nowadays, the biorefinery concept embraces wide definitions
[52,53]. In general, it can be defined as a facility with a range of tech- 5. Life-cycle assessment
nologies able to “break” the biomass into building-block molecules,
which can be converted into value-added products, biofuels, chem- According to the definition of ISO 14044:2006 (www.iso.org),
icals, heat, and power [54,55]. The aim of this bioindustry is to be life-cycle Assessment (LCA) is a tool to assess the potential
competitive in the market and lead to the progressive replacement environmental impacts (natural environment and human health)
of oil refinery products. This concept has been frequently associ- and resources used throughout a product’s/service’s life-cycle. It
ated with environmental concerns. There are three main drivers includes the process from the raw material acquisition, produc-
for the development of biorefinery technologies: climate change, tion and use phases, and waste management such as disposal
energy security, and rural development [55]. and recycling [71–74]. The phases of an LCA study involve goal
Almost all types of biomass feedstocks can be converted into and scope definition, a life-cycle inventory analysis (LCI), life-cycle
different classes of biofuels and biochemicals through conversion impact assessments (LCIA), and interpretation of generated data.
technologies. Biorefineries will most probably encompass a whole The results from an LCA study can be used in many different areas,
range of different-sized installations due to the location of biomass including product creation and development, strategic planning,
resources [55]. Biorefineries can be referred as first (1G), second public policy creation, marketing, and others [71,72].
(2G), or third (3G) generation, according to the substrate used as There are two main different methods for LCA: attributional
raw material [52]. 1G-biorefineries are those that use food crop (ALCA) and consequential (CLCA). ALCA focuses on describing the
resources such as sugar and oil; 2G-biorefineries process non-food environmentally relevant physical flows to and from a life-cycle
raw material such as agricultural residues, wood and energy crops; and its subsystems. Separately, CLCA aims to describe how envi-
3G-biorefineries have been referred to those using algae biomass ronmentally relevant flows will change in response to possible
as a feedstock [52]. decisions [71].
Biorefineries for biochemical production have been looking for LCA studies are more common for poly(lactic acid) (PLA) than
alternative substrates, such as biomass for 2G products. Lignocel- for the lactic acid production chain. The most recent studies about
lulosic biomass is the most abundant source of carbohydrate in the the production of lactic acid from lignocellulosic material, lac-
world, and for this reason it has received a great amount of attention tic acid/ethanol biorefinery and PLA production chain are shown
from biorefineries in the past few years. below.
The lactic acid-based biorefineries may exploit various types Considering the lactic acid production from sugar cane bagasse
of feedstock, such as sugar cane [56,57], sweet sorghum [58,59], and leaves, Gezae Daful and Görgens [42] evaluated a cradle-to-
agricultural food waste [60,61], wastes from bioethanol and beer grave LCA analysis of six different scenarios, as shown in Table 1:
productions [62], municipal organic solid waste [63], corn stover 1-Hemicellulose liquid fraction and Ca(OH)2 ; 2-Cellu-lignin solid
224
Table 2
Literature reports of lactic acid production sorted by higher titer.

Microorganism Product Method Substrate Nitrogen source pH control C Titer Productivity Yield Reference
−1 −1
g/L gL h g/g

Sporolactobacillus sp. CASD D-LA Fed-batch Glucose Peanut meal 5.0–6.0 CaCO3 42 226.00 4.40 0.840 [185]
Bacillus sp. WL-S20 L-LA Fed-batch Glucose Peanut meal 9.0 NaOH 45 225.00 1.04 0.993 [186]
Bacilus coagulans C106 L-LA Fed batch Xylose YE 6.0 Ca(OH)2 50 215.70 4.00 0.950 [100]
Lactobacillus paracasei L-LA Fed-batch Glucose – 6.0 CaCO3 37 215.33 1.79 0.990 [187]
7BL (GMO) Wood chips 99.22 2.25 0.960
Rice straw 66.67 5.27 0.970
Lactobacillus rhamnosus L-LA Fed-batch Defatted rice bran – 6.25 Ca(OH)2 42 210.00 2.56 0.937 [188]
Lactococcus lactis ATCC19435 L-LA Fed-batch Jerusalem artichoke YE 6.0 NaOH 30 142.00 – – [189]

R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239


Lactobacillus casei G-02 L-LA Fed-batch (SSF) Jerusalem artichoke Soybean flour 6.5 CaCO3 40 141.50 4.70 0.963 [190]
Bacillus coagulans XZL4 L-LA Batch Hemp hurds YE 5.5 CaCO3 50 141.00 109.00 – 0.900 0.840 [191]
(Glucose–xylose)
Bacillus coagulans LA204 LA Fed- batch (SSF) Pretreated corncob YE 6.0 NaOH 50 122.99 1.37 0.77 [192]
Lactobacillus agilis LPB 56 L-LA Batch Soybean vinasse – 6.0 Ca(OH)2 30 138.00 0.86 0.849 [193]
Bacillus coagulans LA1507 L-LA Open-fed-batch (SSF) Sweet sorghum Corn steep liquor powder 5.2–6.2 Ca(OH)2 50 111.00 1.59 0.437 [194]
bagasse
Pediococcus acidilactici (GMO) LA Batch (SSF) Detoxified corn stover – 5.5 NaOH 45 104.50 1.45 0.715 [41]
(Cellulose)
Lactobacillus plantarum (GMO) D-LA Batch (SSF) Delignified hardwood – 6.0 NaOH 37 102.30 2.29 0.879 [195]
pulp
Lactobacillus paracasei D-LA Batch (SSF) Curcuma longa waste Soybean meal 6.0 NH4 OH 34 91.61 2.08 0.690 [97]
Lactobacillus coryniformis L-LA 37 97.13 2.70 0.650
Lactobacillus pentosus LA Fed-batch (SSF) Corn stover YE 6.0 37 92.30 1.92 0.660 [196]
Bacillus coagulans JI12 L-LA Batch (SSF) oil palm empty fruit YE 6.0 Ca(OH)2 50 80.60 3.40 – [197]
bunch hydrolysate
Lactobacillus rhamnosus LA Batch (SSF) Recycled paper sludge – 5.5 CaCO3 37 73.00 2.90 0.970 [30]
ATCC7469
Streptococcus sp. LA and biogas Batch Food waste – 6.0 35 66.50 3.38 0.330 [198]
Streptococcus sp. LA Batch (SSF) Food waste YE 6.0 NaOH 35 60.50 2.16 0.810 [199]
Rhizopus oryzae L-LA Batch (SSF) Xylo-oligosaccharides – 5.5 CaCO3 40 60.29 1.00 0.600 [200]
manufacturing waste
Bacillus coagulans L-LA Batch Coffee mucilage YE 6.0 NaOH 52 45.30 4.40 0.770 [103]
Bacillus coagulans L-LA Batch Coffee pulp YE 6.0 NaOH 52 45.30 4.02 0.780 [68]
Lactobacillus paracasei KM2 L-LA Batch Whole slurry of oil YE and peptone 6.0 NH4 OH 37 40.00 – 0.895 [201]
(GMO) palm trunk
Geobacillus stearothermophilus L-LA Batch Raw potato starch YE 7.0 NaOH 60 36.62 1.80 0.660 [202]
DSM494
Lactobacillus delbrueckii LA, xylitol, activated Batch Brewer’s spent grains YE 6.0 NaOH 37 35.54 0.59 0.990 [10,43]
carbon and phenolic (Cellulose)
acids
Lactobacillus pentosus LA Heterofermentation Batch Wheat bran – 6.3 NaOH 30 18.60 0.30 0.730 [67]
DSM20314
Lactobacillus delbrueckii D-LA Batch Cassava fibrous waste YE 6.5 NaOH 37 16.15 0.90 0.500 [203]
delbrueckii NBRC 3202
Lactobacillus casei 12A LA Batch De-oiled algal Amino acids 6.5 37 11.17 – – [66]
biomass + glucose
Bacillus coagulans LA Continuous Non-sterile clarified Corn steep liquor 6.0 NaOH 50 – 3.69 0.950 [26]
corn stover

GMO: Genetically modified organism; LA: Lactic acid; SSF: Simultaneous saccharification and fermentation; YE: Yeast extract.
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 225

fraction and Ca(OH)2 ; 3-Hemicellulose liquid fraction and acid to those of PET. Regarding the LCA, even PLA-based packaging with
tolerant bacteria; 4-Cellu-lignin solid fraction and acid tolerant bac- additives require more energy for their production, so this material
teria; 5-Hemicellulose liquid fraction and Mg(OH)2 ; 6-Cellu-lignin had the highest environmental performance, being both more envi-
solid fraction and Mg(OH)2 . The process included all raw materi- ronmentally friendly and having a lower impact on human health.
als and emissions of sugar cane cultivations, sugar mill, industrial The results still indicated that PLA-based packaging with additives
activities related to auxiliary chemicals, distribution of raw mate- may reduce food loss through extending the shelf-life of the product
rials, processing of sugar cane bagasse and harvest residues for [77].
lactic acid. Scenarios 1 and 2 demonstrated higher environmental In general, the studies for PLA compounds show better envi-
burdens compared to other scenarios in almost all environmental ronmental results when compared to plastic from petrochemical
impact categories, mainly due to the use of Ca(OH)2 and H2 SO4 that sources [75–78]. Furthermore, studies of LCA made specifically for
ends up in the production of gypsum (CaSO4 ) as solid waste. For sce- lactic acid products from large companies [79–81] showed a bet-
narios 3 and 4, the main contributors to environmental impacts are ter performance when compared to petrochemical ones, especially
the nutrients used for the strains, while for 5 and 6 they were tri- by facilitating climate change mitigation and reducing dependence
ethylamine and Mg(OH)2 . In conclusion, the conventional process on fossil and scarce resources while promoting the use of local and
with gypsum as solid waste proved to be the most environmentally renewable resources [78].
unfriendly [42]. Finally, LCA analysis was also used for comparing different
In an evaluation of the impacts of a biorefinery producing ways to dispose the final residues from PLA: chemical recycling,
ethanol from winter wheat-straw and lactic acid production from mechanical recycling and composting. In this case, the method-
alfalfa using CLCA and ALCA approaches, the authors found that ology was used to measure the environmental impact in regards
the best scenario was the integration of the production chains to climate change, human toxicity, and fossil depletion. The LCA
when compared to standalone systems. In the current study, lactic results showed that electricity consumption represents the highest
acid was assumed to be produced from juice fraction and glu- input among mechanical and chemical recycling methods. Mechan-
cose fractions from the hydrolysed mass of the press cake. The ical recycling presented the lowest environmental impact, followed
environmental impacts considered were global warming potential, by chemical recycling and composting. One of the reasons for
eutrophication potential, non-renewable energy use, and the agri- this result is that composting does not produce PLA while the
cultural land occupation. In regards to the LCA, the main benefits other two recycling alternatives produced recycled polymer as an
were in terms of higher net savings of greenhouse gas emissions, output. Therefore, composting the material will be replaced by tra-
non-renewable resources use, and eutrophication potential [70]. ditional production that will cause the impact associated with the
In another case, polymer-grade lactic acid and the ethyl lactate whole production process again. [73]. Similar conclusions were also
production chain from corn stover were evaluated and compared achieved by Hottle et al. [82].
to petrochemical platforms. In reference to LCA results, the author
analyzed life-cycle greenhouse gas emissions and fossil energy con-
sumption. Regarding the end-of-life assumption, the bioproducts 6. Fermentation feedstock for lactic acid production
demonstrated lower greenhouse gas emissions (23–90%) and lower
life-cycle fossil energy consumption (10–72%) than fossil-derived Fermentation feedstock represents a considerable part of the
compounds [75]. production costs in the lactic acid production process, together
Considering PLA production chain, Gruber [76] analyzed the with nutritional requirements for the chosen fermentation micro-
impacts compared with other plastics using an LCI methodol- organism [40,44,45,83]. Industrially, the most used genus of
ogy. PLA production was considered, from corn to 1 and 5 years micro-organisms in the production of lactic acid is Lactobacillus sp.
after the start of operations at the plant, and in the long-term. As It possesses specific nutritional needs, such as nitrogen, vitamins,
expected, the amount of fossil fuels needed and the amount of CO2 and minerals. These nutrients are used in cellular growth mainte-
emissions from PLA production is significantly lower than other nance and lactic acid production, which are in fact associated.
plastics, tending to further reduce the plant operating time. In addi- There are two main bottlenecks to produce lactic acid by micro-
tion, they compared the disposal of products made from PLA and bial fermentation [84]. The first one is related to the high costs of
polystyrene in terms of required fossil fuels and CO2 emissions. The sugars used as a substrate. The second one is due to the cost of
fermentation of lactic acid and PLA production is an extremely effi- sterilization (thermal treatment) and downstream processes (sep-
cient process, which is essential for a process to become desirable aration and purification steps) [85]. Knowing these challenges, it
and economically viable, especially for products from renewable is necessary to improve the production process on issues, such
resources. In all cases reported by the author, the PLA possessed as: expensive micro-organism nutritional requirements, carbon
advantages compared to other plastics, since it reduced energy con- source cost, controlling the acidity of the medium that affects
sumption and CO2 emissions in the process, as well as allowing for fermentation, and controlling the process to minimize product
a wider range of disposal processes since it is biodegradable. PLA is inhibition impact. Overcoming all these barriers means achieving a
highly stable under normal conditions, but degrades rapidly when higher lactic acid production with lower costs [86], once the costs
exposed to environments with high temperatures, humidity, and of the substrates cannot be overcome just by scaling-up [87].
high microbial activity. This allows alternative disposal processes, However, there are some options of feedstock that have been
such as compositing and anaerobic digestion [76]. tested for cost reduction in lactic acid fermentation, such as cas-
In the specific case of using PLA for fruit packing, the sava starch [19], cornstarch [88], fresh sweet potato [89], sugar
authors used an LCA methodology in order to compare PLA- cane molasses [2], Zizyphus oenophlia biomass [90], white rice bran
nanocomposite (nano-clay and surfactant) packaging, pristine PLA [91,92], corn stover [84,93,94], sugarcane bagasse [2,95], coarse rice
and polyethylene terephthalate (PET). The analysis focused in eval- [96], Curcuma longa biomass [97], paper sludge [98], green algae
uating physiochemical and antimicrobial properties, as well as [99], xylose [100], inulin from Jerusalem artichoke [101], mango
the environmental impacts regarding the raw material acquisi- peel [102], coffee residues [68,103], and whey [104]. Despite the
tion, production of materials, packaging manufacture, filling of wide variety of feedstock tested for lactic acid production, and
packaging and end-of-life solutions. The results showed that PLA- even presenting some promising results, most of them have prob-
nanocomposite displayed a better performance, thus contributing lems with price, seasonality, fermentation rate, yields of lactic acid
to bringing the characteristics and behaviour of PLA packages close production, and the presence of by-products [5,105].
226 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

Carbon sources currently used industrially for the production of Given all this, it is clear that, although lignocellulosic biomass
lactic acid consist of carbohydrates such as glucose, lactose, starch, is suitable for the production of lactic acid, this process is still
maltose, and sucrose obtained from sugar beet, molasses, whey, and expensive and not fully understood. For this reason, there is still
barley malt [5]. These substrates are already processed, which ends much to develop in the area in order to make 2G-lactic acid eco-
up endearing the process [19], as well as competing with the food nomically viable on a commercial scale. Some examples of recent
supply. Comparative results of different fermentation processes studies using lignocellulosic materials for lactic acid production are
for lactic acid production presented in the literature are shown in presented in Table 2.
Table 2.
6.3. Microalgae
6.1. Starchy materials
Algae biomass is also a suitable substrate for lactic acid pro-
There are a lot of starchy materials that can be used for lactic acid duction. It has a high content of carbohydrates and proteins, and
production, such as: corn, maize, rice, rye, wheat, potato, barley compared with lignocellulosic biomass, presents the advantage
and cassava [106]. In general, these substrates work very well in of not having lignin [16,115,116]. As an example, Hydrodictyon
lactic production, especially when associated with amylolytic lactic reticulum contains 47.5% of polysaccharides. This material can be
acid bacteria [22,107]. Their use also allows micro-organisms to converted to glucose and mannose as fermentable sugars with-
grow without presenting substrate inhibition, generally caused by out producing as many inhibitory compounds as in lignocellulosic
glucose [106]. However, their use presents a well-known problem, biomass processing [115,116]. The use of microalgae and cyanobac-
since these materials are used for human and animal feeding. The teria as lactic acid producers could also reduce the feedstock cost,
competition of the lactic acid industry with the food sector is the due to their ability to use light energy to fix CO2 [16,117]. An exam-
same already demonstrated for the biofuels sector [108]. ple is presented in Table 3, using Synechocystis sp.
In summary, the main advantages of using microalgae as
6.2. Lignocellulosic biomass biomass for producing 3G-lactic acid are the facts that it is
possible to grow everywhere with short harvesting times, they
Currently, lignocellulosic biomass is the most promising sub- do not present lignin content, which simplifies saccharification,
strate for lactic acid production. This kind of biomass is the most and they have high fermentable sugar contents and proteins
abundant material that can be used for lactic acid production [16,87,115,117]. Table 2 presents an example of algae biomass as a
around the world. It comprehends all green biomass and a great feedstock for lactic acid production.
amount of agro-industrial residues. These materials present the
main advantage of not competing with the food supply. Besides 6.4. Food waste
that, lignocellulosic materials from agricultural, agro-industrial,
and forestry sources are a potentially inexpensive and renewable Food waste can be any material from the food supply chain, from
carbohydrate feedstock for large-scale lactic acid fermentation. the production process to the wastes generated by the final con-
These materials are abundant, low price, contain a high polysac- sumer. Food waste presents a high carbohydrate content [16], also
charide content, and are renewable [33]. making it suitable as a substrate for lactic acid production. Sev-
Lignocellulosic materials from agroindustry include a variety eral studies revealed these kitchen residues/refuse are suitable for
of materials such as sawdust, poplar trees, sugar cane bagasse, many processes for lactic acid production such as mixes of cooked
waste paper, brewer spent grains, switch-grass, and straws, stems, rice, vegetables, meat, and bean curd [118]; rice, noodles, meat,
stalks, leaves, husks, shells and peels from cereals like rice, wheat, and vegetables [38,119]; vegetables such as carrot peel, cabbage,
corn, sorghum and barley, among many others [109]. However, the and potato peel, fruit such as banana peel, apple peel, and orange
cellulose and hemicellulose in the lignocellulose material are not peel, baked fish, rice, and used tea leaves [120,121]; rice, noodles,
directly available for bioconversion to lactic acid, because of their meat and vegetables, and unsold bakery products including cakes,
intimate association with lignin and lack of hydrolytic enzymes in breads and pastries [64]; rice, vegetables, and meat [122,123]; cof-
the most common micro-organisms [33]. fee mucilage [103]; and coffee pulp [68]. Some results of processes
The process of breaking down polysaccharides into consum- using food waste materials for lactic acid production are depicted
able sugars can start with a pretreatment process to break the in Table 2.
lignocellulose structure, followed by an enzymatic hydrolysis to
de-polymerize lignocellulose to fermentative sugars [106]. The 6.5. Glycerol
amounts of carbohydrate polymers and lignin obtained vary
between plants and species. Also, the ratios between the sugars Glycerol, as a by-product of biodiesel production, can be used as
in a single plant may also vary with age, stage of growth, and other a substrate for lactic acid production. Biodiesel production has been
conditions. The separation of the main fractions present in lignocel- increasing every year and the glycerol yield is about 10% of biodiesel
lulose biomass is of great interest, since the fermentation process production [115]. Since biodiesel is considered an environmentally
will subsequently be performed. Also, by-products obtained during friendly fuel [87], the possibility of using glycerol residue for pro-
pretreatment hydrolysis of these materials have to be considered, ducing lactic acid increases its visibility in the global market even
since they may affect the fermentation yield [109–112], and some- more. Furthermore, it is important to consider that a great diver-
times even change the metabolic pathways of the cells [113]. sity of vegetable and algae is used to produce biodiesel around the
The pretreatment step for breaking the lignocellulosic struc- world, which makes the overproduction of glycerol as a by-product
ture generates a range of side-products that act as inhibitors for a challenge still to be overcome [87]. Reports indicate that some
the ensuing process of enzymatic hydrolysis and also for fermen- micro-organisms are able to directly consume glycerol to produce
tation [113,114]. The inhibitors can be grouped into three major lactic acid [124–126]. However, it is still a challenge to make this
categories: weak acids such as formic (from furfural degradation), conversion without using a genetically modified micro-organism,
levulinic (from HMF degradation), and acetic (formed by the de- or even without additional components to balance cell redox poten-
acetylation of hemicellulose); furans as 5-hydroxymethylfurfural tial. For this reason, it is considered an alternative to use glycerol
(HMF) and furfural (from pentose and hexose dehydration); and with a coupled metabolism of another substrate with higher oxi-
phenolic compounds (mainly from lignin breakdown) [114]. dizing power to restore the redox balance [124]. Nevertheless, as
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 227

Table 3
Literature reports using genetically modified micro-organisms to produce lactic acid.

Microorganism Modification Improved phenotypic Result Reference

Kluyveromyces L-lactate dehydrogenase (LDH) expression of L-lactic acid production from 24.0 g/L, yield of 0.48 g/g [204]
marxianus Staphylococcus epidermidis, Lactobacillus glucose
acidophilus, and Bos taurus
Saccharomyces Expression of cellodextrin transporter (CDT-1, Lactic acid production from yield of 0.358 g/g [205]
cerevisiae which also transports lactose) and a lactose
␤-glucosidase (GH1-1, which also acts as a
␤-galactosidase) from Neurospora crass.
Expression of lactate dehydrogenase (LDHA)
from Rhizopus oryza
Saccharomyces Expression of D-lactate dehydrogenase gene D-lactic acid production at low 82.6 g/L, yield of 0.83 g/g, [206]
cerevisiae (LDHA) from Leuconostoc mesenteroides. pH productivity of 1.50 g/L/h at pH
Deletion of genes: ADH1, ADH2, ADH3, ADH4, 3.5
ADH5, GPD1, GPD2 and DLD
Saccharomyces Expression of D-lactate dehydrogenase (LDHA) D-lactic acid production 112.0 g/L, yield of 0.80 g/g, [207]
cerevisiae gene from Leuconostoc mesenteroides subsp. productivity of 2.2 g/L/h
mesenteroides ATCC 8293. Deletion of genes:
DLD1, JEN1, PDC1, ADH1, GPD1 and GPD2.
Overexpression of HAA1
Saccharomyces Expression of 12 glycolysis-related genes and D-lactic acid production 60.3 g/L, yield of 0.646 g/g, and [208]
cerevisiae D-lactate dehydrogenase (DLDH) from productivity of 2.80 g/L/h
Leuconostoc mesenteroides
Monascus ruber Introduction of lactate dehydrogenase (LDH); Lactic acid production at low 190 g/l at pH 3.8 and 129 g/l at [209]
two genes encoding pyruvate decarboxylase pH pH 2.8
(PDC) were knocked out; 4
cytochrome-dependent LDH (CLDH) genes
were knocked out
Escherichia coli Replacement of D-lactate dehydrogenase gene L-lactic acid production from 75 g/L, yield of 85%, [210]
(LDHA) by L-lactate dehydrogenase gene molasses and corn steep liquor productivity of 1.18 g/L/h, >99%
(LDHL) from Pedicoccus acidilactici; Enhanced without additional nutrients optical purity
expression of the sucrose operon (cscA and
cscKB)
Synechocystis sp. Increasing the expression level of lactate L-lactic acid production from 12.99 ± 3.3 g/L, and carbon [211]
dehydrogenase (LDH), co-expression of a CO2 partitioning into lactic acid of
heterologous pyruvate kinase, knockdown of 50.0 ± 5.9%
phosphoenolpyruvate carboxylase, and
optimization through site-directed
mutagenesis to improve the enzyme’s affinity
for the co-factor nicotinamide adenine
dinucleotide phosphate (NADPH)
Lactobacillus plantarum Deletion of L-lactate dehydrogenase (LDH), D-lactic acid production from 38.6 g/L [212]
phosphoketolase (xpk1) gene substituted by a arabinose
heterologous transketolase (tkt) gene from
Lactococcus lactis
Lactobacillus plantarum Deletion of L-lactate dehydrogenase gene D-lactic acid production from 73.2 g/L, yield of 0.85 g/g, [213]
(LDHL1), expression of alpha-amylase (AmyA) raw corn starch optical purity of 99.6%
from Streptococcus bovis
Lactobacillus plantarum Deletion of L-lactate dehydrogenase (LDH), D-lactic acid production 27.3 g/L and productivity of [214]
expression of xylose isomerase and through simultaneous 0.75 g/L/h from corn stover;
xylulokinase genes from Lactobacillus pentosus saccharification and 22.0 g/L and productivity of
fermentation from xylose 0.55 g/L/h from sorghum stalks

innovative technology approaches, it needs to be well developed in although not all micro-organisms are able to consume it directly.
order to be made suitable for industrial scale . Additionally, reuse of waste is of great interest due to legislation
and environmental issues, since the industry is increasingly being
6.6. Feedstocks concluding remarks induced to find an alternative to the use of waste material from its
production [131].
Research efforts have been increasingly focused on finding new Moreover, multiple carbohydrates have been used in the pro-
and effective nutritional sources, always associated with new fer- duction of lactic acid, but the substrate used depends directly on
mentation techniques. It reflects the attempt to implement these the location of the lactic acid production plant, due to both avail-
processes in industrial terms with a high substrate conversion ability and logistics of transportation and use [33]. Furthermore,
and high lactic acid production [127]. In fact, the application of there is a difficulty related to the fact that many substrates gen-
agro-industrial residues in bioprocesses provides an alternative to erate complex biological process which culminates in high cost of
minimize, or even replace, refined and expensive raw materials recovery and purification of lactic acid [5].
such as glucose and also helps to reduce environmental risks from
these waste [102].
Currently, the use of low-cost waste materials in the industrial 7. Lactic acid bacteria
production of lactic acid is still rare [102]. Lignocellulosic biomass
is expected to be a cheap and renewable carbon source [3,128,129]. Lactic acid bacteria are a very heterogeneous group in terms
Moreover, it does not have a direct value as food [5] allowing of classification, often suffering phylogenetic relocation. Gen-
the use of inexpensive, abundant and renewable carbon sources eral descriptions of the group include characteristics, such as:
[130]. It can be a good substitute for conventional raw materials, gram-positive, non-sporing, catalase negative, non-aerobic but
228 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

aero-tolerant, fastidious, acid-tolerant, which produce lactic acid capital and operation investment, since it is possible to share some
as the major end product during the fermentation of carbohydrates facilities and personnel.
[132]. They are highly adaptable, and it is already well known that
they have a great metabolic diversity. They can survive in a wide 7.2. Choosing micro-organisms – desirable characteristics of
range of environmental conditions and the only feature that cov- Lactobacillus sp.
ers the whole group is being gram-positive [132]. 90% of studies
on lactic acid production are related to lactic acid bacteria, which Although many micro-organisms produce lactic acid, the most
are related to high yields, productivity [11], and its environmental commonly used by the industry is Lactobacillus sp. [93]. They have
versatility, covering a pH range from 3.2 to 9.6, and temperatures a high tolerance to acidic environments and are easily genetically
from 5 to 45 ◦ C [133]. modified for selective production of lactic acid isomers [149]. More-
For the industrial production of lactic acid, it is desirable that over, these micro-organisms are considered safe for the industrial
the micro-organisms used are able to ferment cheap raw mate- production of lactic acid, due to their long history in industrial pro-
rials, have low nitrogen requirements, produce large amounts of duction in many sectors with no reports of adverse effects, both for
lactic acid, tolerate low pH conditions and high temperatures, and workers and for consumers of products [87].
produce the least amount of by-products [20]. The main fermenta- When used for industrial purposes, it is desirable that micro-
tion micro-organisms used nowadays are homofermentative from organisms present some general characteristics that facilitate their
the genus Lactobacillus, Streptococcus, and Pediococcus [37]. Some handling and increase the profitability of the process. As shown
examples from literature are shown in Table 2. Considering the in Table 2, Lactobacillus sp. represents the majority of micro-
importance that the genus Lactobacillus has for lactic acid produc- organisms studied for lactic acid production, due to its beneficial
tion, the following sections will focus on the metabolism of these characteristics for the process.
micro-organisms. Micro-organisms should have a high conversion efficiency of
substrate to product. At the same time, they should allow product
7.1. Nutritional requirements for lactic acid bacteria accumulation in the broth (that means they do not suffer by-
product inhibition), since these factors impact on the final cost of
Another important issue to be considered, when it comes to the the product [83]. It is also necessary to consider the time spent dur-
association with substrates for fermentation and lactic acid bacte- ing the process by the micro-organism to convert substrate into
ria, is the high nutrient requirements of these micro-organisms. In product (productivity), since it will have a direct impact on the
general, besides being a source of carbon used in energy generation, infrastructure of the project. So, making an efficient conversion and
lactic acid bacteria requires nitrogen and a wide range of vitamins a high concentration of the product is essential, in order to reduce
and minerals to maintain cellular growth and lactic acid production. recovery costs, among others, since they can represent up to 50%
It occurs because they have a limited ability to synthesize molecules of the total process cost [46]. It is important to consider that the
for their own growth [11]. micro-organism should not produce metabolites that are incom-
The ratio between nitrogen and carbon sources is one of the patible with the product of interest, as this would jeopardize the
main factors affecting the sugar conversion yield into lactic acid viability of the process. To assure the safety and stability of the
[11]. This situation may be overcome by the addition of complex production pathway, micro-organisms should not be pathogenic,
nitrogen sources in the fermentation medium. Yeast extract is com- ensuring the safety of workers, consumers and environment. They
monly used, as well as peptone and meat extract. In particular, lactic have to be physiologically stable so the process behaviour can be
acid production gets better results when yeast extract is added to predicted, operated and easy to handle. Growing conditions should
the medium [11]. However, this addition greatly increases operat- be simple and the cultivation broth should be inexpensive, ensuring
ing costs. For this reason, much effort has been focused on trying to the economic viability of the process [83].
find alternative sources of nitrogen, minimizing costs without loss
in product yield. 7.3. Metabolic pathways for lactic acid production by
Until the mid-90s, the vast majority of studies concluded that Lactobacillus sp.
yeast extract was an irreplaceable source of nitrogen when com-
pared to alternative sources such as corn steep liquor, casein An important subject to be considered in the production perfor-
hydrolysate, hydrolyzed whey protein, among others [134,135]. It mance of lactic acid is the metabolic pathway used by the chosen
was associated with the fact that yeast extract is an excellent source micro-organism. There are essentially two pathways in which lactic
of B vitamins and is often used to provide these factors in bacterio- acid bacteria can produce lactic acid. According to these pathways,
logical culture media [136]. Since then, it is considered essential to the micro-organisms can be classified as either homofermentative
achieve rapid rates of cellular growth and lactic acid production by or heterofermentative bacteria.
lactic acid bacteria [136,137]. However, studies have shown better The homofermentative uses the glycolytic pathway
results in replacement of yeast extract for alternative and cheaper (Embden-Meyerhof-Parnas pathway). Therefore, the final
nitrogen sources, such as malt extract, white rice bran hydrolysate, product is almost exclusively lactic acid (Fig. 2A) [143]. On
corn steep liquor, and malt sprouts [91,138–142]. Some examples the other hand, heterofermentative micro-organisms use 6-
are shown in Table 2. It has to be highlighted that the costs of phosphogluconate/phosphoketolase pathway (6-PG/PK), which
these inputs are region dependent. For example, in Brazil, yeast results in other by-products in addition to lactic acid, such as
extract could still be an important alternative if lactic acid pro- ethanol, acetate and CO2 (Fig. 2B) [132]. The crucial difference
duction could be carried out as an annex to a sugar, ethanol and between these metabolic pathways is given by the presence of key
yeast production plant. It is important because it is believed that enzymes for each route [132]:
the good results in lactic acid production may be a consequence
of the choice of complex sources which contain soluble proteins, • Fructose 1,6-diphosphate (FDP) aldolase to glycolysis pathway;
amino acids, B vitamins, and other nutrients available to the micro- • Phosphoketolase to 6-PG/PK pathway.
organism. At this point, it is worth mentioning that building up
2G-lactic acid facilities in a 1G-sugarcane ethanol facility may also Strictly homofermentative micro-organisms produce lactic acid
be quite interesting since sugar cane bagasse is readily available on from glucose using the glycolytic pathway, thus they are not able to
the production site, reducing the logistical costs as well as the total metabolize pentose sugars and related compounds. The heterofer-
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 229

Fig. 2. Metabolic pathways for lactic acid production: (A) Embden-Meyerhof-Parnas pathway (EMP); (B) 6-phosphogluconate/phosphoketolase pathway (6-PG/PK).

mentative bacteria group produce lactic acid and other compounds


using 6-PG/PK pathway from hexose and also from pentose sugars
[144].
However, facultative heterofermentative bacteria metabolize
glucose by the glycolytic pathway, but pentose metabolism occurs
by the 6-PG/PK pathway. In this case, the production of lactic acid
can or cannot be associated with the production of other com-
pounds of 2 carbons, depending on the micro-organism [144].
These micro-organisms have the enzymes needed to work in
both the above-mentioned metabolic pathways. Thus, such micro-
organisms use FDP aldolase to ferment hexose, but in the presence
of pentose, they are induced to produce phosphoketolase. This
means that the presence of hexose results in an homo-lactic
fermentation, whereas pentose can result in hetero-lactic fermen-
tation [132].
In the case of the facultative heterofermentative micro-
organisms, what determines the behaviour to lead to homo or
heterofermentation can either be the source of carbon or the expo- Fig. 3. Metabolic pathways using xylose for the production of lactic acid.
sure to certain environmental parameters, such as nutritional,
osmotic and/or thermal stresses, depending on the micro-organism If one continues to consider the sugar supplement, it is impor-
species. tant to consider that some Lactobacillus sp. suffer suppressed
In fact, many lactic acid bacteria are able to consume pen- growth due to the excess of monosaccharides, since this can gen-
tose molecules as arabinose, ribose, xylose and carbohydrates as erate excess glucose in the cell [145]. To avoid this condition, the
gluconate. Note that xylulose-5-P are present in both heterofer- micro-organism releases glucose to the medium, spending energy
mentative (Fig. 2B) and xylose assimilation pathways (Fig. 3), that could be used in the production of lactic acid. Thus, the excess
allowing pentose sugars to be consumed by any of these routes of fermentable sugar is a major drawback in the industrial produc-
depending on the chosen micro-organism. tion of lactic acid.
The theoretical value of the lactic acid production by pentose Another important issue to be considered when it comes to
molecule is 1.67 in homolactic fermentations (3 xylose molecules to lactic acid bacteria is the high nutrient requirements for these
generate 5 molecules of lactic acid, totalling 15 carbons) (Fig. 3). In micro-organisms. All lactic acid bacteria have high nutritional
this case, the pentose-phosphate metabolic pathway is used, with requirements due to their extremely specific environments and low
the action of transketolase and transaldolase enzymes [144]. ability to synthesise amino acids [146]. In general, besides a source
230 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

of carbon used in energy generation, these bacteria require nitro- changes in the cytoplasmic membrane, making them more com-
gen, plus a wide range of vitamins and minerals for the maintenance pact and rigid [152,153,158]. They all corroborate that this theory
of cellular growth and to support the lactic-acid-releasing mecha- is widely accepted for different species of Lactobacillus sp.
nism. This occurs because they have a limited ability to synthesize Besides acid stress, there are many other factors that influ-
molecules for their own growth [11]. It also probably happens ence the survival of lactic acid bacteria. Among the main ones, the
because these micro-organisms have gone through an evolution most emphatic are nutritional stresses, temperature, oxidative and
process associated with rich media, such as meat and milk. It pre- osmotic stress, and the presence of chemicals.
sumably prevented them from developing/keeping cell machinery Environmental changes are sensed by the bacterial cell through
that synthesizes some molecules for growth, once it was present in the translational signal system, which is responsible for adap-
the substrate. For this reason, these bacteria have difficulty to cap- tive cell changes in metabolism, physiology, and behaviour [159].
ture inorganic nitrogen from the medium. However, they are quite This system sends signals to the various cellular mechanisms each
efficient at breaking down complex proteins [144]. For this reason, time that an environmental change is detected. This system has
they are dependent on the presence of peptides and amino acids in two main components: the sensor membrane and the cytoplas-
the culture medium [145]. mic response regulator. Based on this model, Zhai et al. [153] were
Acetate is another nutritional factor present in almost the whole able to detect changes in both protein and transcription levels for L.
culture medium. Some researchers suggest that the presence of delbrueckii bulgaricus since the cells respond quickly to the delete-
sodium acetate is critical in the production of a particular isomer of rious effects of acid stress using molecular rapid responses to repair
lactic acid [147]. For example: in absence of acetate, Lactobacillus damage and allow for survival.
sakei produces a racemic mixture of lactic acid [147]. Also, sodium Peptidases over-expression was observed in L. casei [160] and in
acetate has an inhibitory effect on the growth of fungi [148] and L. rhamnosus [157]. The authors believe that this happens because
other contaminant lactic acid bacteria, allowing the growth of Lac- the intracellular amino acids become limited, since the efficiency
tobacillus in general. of amino acid transporters is at a reduced low pH. In this case, the
cell is forced to find alternatives to their own amino acid supply
7.4. Main metabolic characteristics of Lactobacillus sp. under through the over-expression of proteins that assist in the transport
stress conditions of peptides and peptidases that help in the degradation of misfolded
proteins to recycle amino acids [153].
There are several factors that must be considered when It is already known that nutritional changes can cause big
discussing the industrial production of lactic acid by these micro- impacts on lactic acid bacteria [147]. In the previous section, the
organisms, since they are always subjected to stress conditions role of different carbon sources in the metabolic pathways of lac-
during the fermentation process. tic acid production by lactic acid bacteria was mentioned. But, not
Product inhibition can be caused by several factors, such as: the only different types of sugars affect lactic acid production. Other
collapse of cell membrane due to the change of membrane poten- components such as inorganic salts, acids and peptides as well as
tial, anions accumulation inside the cells, high concentration of their concentrations also have major impacts on both lactic acid
solutes, or even acidification of cytosol [130,150,151]. An important production and productivity.
example is the acid stress condition caused by lactic acid. Although In low glucose concentrations, L. plantarum metabolism is
lactic acid bacteria have cellular mechanisms of protection against shifted to acetate production, and not to lactic acid (Fig. 4). This
a pH drop, they suffer inhibition caused by the product, since this happens due to the need to produce at least one ATP to maintain
phenomenon is caused by the cell’s own lactic acid production. cellular homeostasis [161]. This process begins with the conver-
In gram-positive bacteria, acid stress directly affects the ATP sion of lactate to pyruvate by lactate oxidases (LOX). Pyruvate is
available in the cell for proton pump maintenance [152]. It results in then converted to acetate in order to generate ATP. This mecha-
a reduction of energy available for the synthesis of biomass, and also nism can occur via three different routes: pyruvate formate lyase
for the catabolic flux of the micro-organism. Although the mecha- (PFL), pyruvate dehydrogenase complex (PDH), or pyruvate oxidase
nism is not fully explained, it is known that such disturbances alter (POX) (Fig. 4).
gene transcription of the central metabolism of the cell, which is The first one may occur under anaerobic conditions by pyru-
sensed due to the increased presence of proteins. These changes vate formate lyase (PFL), phosphotransacetylase (PTA) and acetate
are not noticed during a fast drop in pH, possibly due to an increase kinase (ACK), resulting in the production of formate and acetate.
in the stability of mRNA. But, since the environment is acidic, there However, Lorquet et al. [161] highlighted that it was not possible
is a substantial increase in glycolytic enzymes. This process allows to detect formate during acetate production.
the cell to be prepared for fast growth recovery and normalization The second possible route occurs via pyruvate dehydrogenase
of metabolism when pH stress is removed. complex (PDH), PTA and ACK. In this way, NAD+ is converted in
Zhai et al. [153] used L. delbrueckii bulgaricus to demonstrate the NADH + H+ . This does not occur in the presence of O2 because in this
consequences of acid stress to cells. The pH used in the tests was 3.7 case there are already high levels of NADH in the cell. Furthermore,
which is quite close to the pH reached in lactic acid fermentations since there is no transformation of pyruvate to lactate, NAD+ levels
without neutralization (around 3.8) [154]. In the case of acid stress, fall, since NADH recycling does not occur. This further complicates
they showed that the pyruvate from a sugar breakdown is shifted the use of this route.
to the production of acetyl-CoA and not to further lactate produc- Finally, the highest yields of acetate are given in aerobic condi-
tion. This deviation allows the acetyl-CoA to be diverted to fatty tions [161–165] and glucose limitation, by reaching a homoacetic
acid biosynthesis, which modifies the fluidity of the cell membrane fermentation. The proposed route for this process uses POX and
[153,155]. It makes the membrane more resistant to trades with ACK. In this pathway, there is a final production of acetate, CO2 ,
the external environment, achieving energy savings for the cell in and an ATP molecule, with no use or recycling of NAD+ . The activa-
the transfer of protons [153], while all ATP are converted to proton tion of this pathway is made by a group of genes called pox, which
motive force maintenance. Similar processes were observed for L. are responsible for producing the enzyme POX. The gene poxB is
casei [156] and for L. rhamnosus [157]. It was also suggested that mainly responsible for producing the enzyme and is therefore the
acid stress leads to the production of one more ATP synthase [155]. target of several studies. It is known that this group of genes can be
Other studies corroborate this fact, revealing that exposure to acid activated by three factors: low glucose concentration, and the pres-
stress leads bacteria to counteract the influx of protons through ence of O2 and/or H2 O2 . When glucose is available, but the gene is
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 231

Fig. 4. Lactate and acetate production pathways in L. plantarum [161].

activated by O2 or H2 O2 , the productions of acetate and lactate are are from the functional categories related to transportation and
concomitant. Oxygen is required as a substrate for the POX enzyme carbohydrate metabolism, energy metabolism, stress responses,
and it strongly induces transcription from the poxB promoter by an nucleotide and nitrogen metabolism, and biosynthesis of proteins.
unknown mechanism [161]. Thus, depending on the selected strain, L. plantarum is able to
An interesting fact is that since the pathway LOX-POX-ACK metabolize 15–25 different carbon sources. In relation to the car-
stops working, the biomass is degraded by cell lysis. This happens bohydrate metabolism, osmotic stress caused by high levels of
because, in the absence of ATP, protons cannot be removed and cell carbohydrates inhibits glucose metabolism and induces alterna-
homeostasis is disrupted by dissipation of the proton motive force. tive mechanisms. Thus, they showed that the highest number of
Apparently, this is an autolysis mechanism that occurs in gram- carbon sources metabolised by L. plantarum occurs when cells are
positive bacteria [152]. A product of the LOX-POX-ACK pathway pre-cultured in medium with little or no glucose.
acts as a messenger to regulate cell death and lysis in L. plan- These findings corroborate with carbon catabolite repression
tarum, since a strong correlation between the acetate production (CCR), which is a hierarchical system of carbon consumption. Thus,
rate and cell viability is established [162]. It was found that the by a complex regulatory system, the consumption of glucose leads
slower lactate was converted to acetate, the longer cells retained to a suppression of the synthesis of enzymes that are related to
their viability. Besides this, more acetate was produced than lac- the use of other carbohydrates [155]. Thus, it is concluded that the
tate was consumed, resulting in a carbon balance greater than one expression of proteins responsible for input and metabolism of car-
[162]. The authors suggested that the additional acetate could be bohydrate is highly dependent on the carbon source present in the
produced at the expense of other compounds present in the growth culture medium. It is further considered that hostile environments
medium, such as serine catabolism, which seems to be associated such as low pH, high level of carbohydrates, and buffering capacity
with acetate formation [146,166]. In this case, serine is converted negatively affect the kinetics and growth of this micro-organism
into ammonia and pyruvate, which is degraded to form acetate and [155].
other compounds, depending on the strain under study. According Iino et al. [147] tested about 50 different lactic acid bacteria.
to Liu et al. [166], L. plantarum cells in the stationary phase convert Although the authors showed that L. plantarum is a resistant bac-
serine to ammonia, acetate and CO2 , which could be an explanation teria to applied environmental variations, there are some factors
for the carbon balance greater than one. that shown to be more effective for obtaining higher concentration
Using proteomic techniques, it was demonstrated that pro- and productivities during lactic fermentation. The effects of pres-
tein expression from L. plantarum depends on both the medium ence/absence and concentration of salts and other components in
composition and the strain used [155]. The authors reported that the culture medium for L. plantarum are highlighted since many of
the main proteins regulated by the composition of the medium them directly affect the lactic acid production [147]:
232 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

• Carbon Source: Carbon sources such as arabinose and ribose leading to improvements of important strains, such as the ones
resulted in a production of around 50% of acetic acid and 50% used in the food industry.
of lactic acid. The highest lactic acid production occurred using The Lactobacillus genus represents the largest and most diverse
sucrose; genus of all the lactic acid bacteria. These micro-organisms are easy
to genetically modify and present a great capacity for adaptation to
• Organic salts: Presence of salts from organic sources resulted in new environmental conditions. The increasing number of available
an increase of lactic acid production; Lactobacillus sp. genome sequences has allowed for understand-
• Sodium acetate: Presence of sodium acetate in the broth resulted ing of the genetic and metabolic potential of this group. They are
in an increase of 45% in production of lactic acid; recognized as potential cell factories, confirmed by the successful
• Temperature: The best temperatures to produce lactic acid were production of many compounds [168].
around 30 ◦ C–37 ◦ C. Although Ghaffar et al. [5] had mentioned Some examples of genetic modification of strains to produce lac-
43 ◦ C for L. plantarum, many studies used temperatures of around tic acid are shown in Table 3. In the most recent studies, yeasts have
30 ◦ C–37 ◦ C; a central role in the development of new potential micro-organisms
• Oxygen presence: A decrease in lactic acid production and an for lactic acid production, especially dealing with low pH in organic
increased production of concomitant acetic acid in aerobic cul- acid fermentation. The reason behind this is that acid stress and
tures was observed compared to stationary cultures (35% of acetic end-product inhibition are still among the greatest bottlenecks in
acid production in aerobic cultures); commercial lactic acid production, and the heterologous expres-
sion of enzymes in yeasts and fungi renders them more tolerant to
Bron et al. [164] used the fractional factorial design to study acid than lactic acid bacteria [167].
variables on the metabolism of L. plantarum in producing lac- Additionally, genetic engineering is also focusing on the pro-
tic acid, such as temperature, pH, O2 , amino acid concentration, duction of one specific lactic acid isomer. The most recent studies
and the presence of NaCl. The results were focused on studies of are concentrated on the expression of lactate dehydrogenase (LDH)
ODmax (maximum optical density for biomass yield), ␮max (maxi- enzymes in micro-organisms that naturally do not contain the gene
mum growth rate), and the whole transcriptome of expressed genes for lactic acid production, in order to achieve high D- or L-lactic acid
influenced by the parameters analysed. Its results corroborate with concentration and productivity.
all results previously presented for this micro-organism: As raw material is a major cost factor in lactic acid production,
many efforts have been directed towards the utilization of lignocel-
• The ␮max was observed at the temperature of 37 ◦ C and pH of 5.2; lulosic biomass and biomass-derived waste streams [167]. Genetic
• The concentration of amino acids had no influence in the process. engineering efforts in this direction were very well depicted in the
The authors suggest that when glucose is a limiting factor the work of Mazzoli et al. [169], presenting recombinant lactic acid
addition of amino acids will not have a great impact; bacteria with improved amylolytic, cellulolytic or hemicellulolytic
• The production of acetate was substantially higher in the pres- properties. In Table 3, Lactobacillus plantarum is the main example
ence of O2 ; this is explained by the conversion of lactate to acetate of a micro-organism with genetic modification directed towards
aerobically [162,163]; substrate consumption.
• The ODmax was considerably lower in the presence of NaCl; New technologies such as the CRISPR/Cas system represent a
• The conversion of citrate to succinate depends on NaCl presence. new advance for all areas related to micro-organisms. CRISPR (clus-
This conversion is associated with a decrease in OD. It is likely tered regularly interspaced short palindromic repeats), together
that lactate degradation cascade generating an additional ATP is with CRISPR-associated genes (cas) form a bacterial immune sys-
non-functional in the presence of NaCl. In addition, salt presence
works altering the osmotic pressure of the cell, decreasing citrate
transport efficiency through the membrane.

The transcriptome analysis showed that genes associated with


the responses tested are conserved genes within the L. plantarum
sub-species [164]. This allows us to assume that this type of
response presents similarities independent of the strain used. The
authors also assert that the tested parameters associated with
physiological parameters increase the probability that these results
can be extrapolated with other culture media, conditions and
scales.

7.5. Genetically engineering lactic acid bacteria

Genetically engineered lactic acid bacteria have been widely


studied in the last few years. The major challenges associated with
lactic acid production that can be addressed by employing genetic
engineering strategies are isomer purity, micro-organism acid tol-
erance, carbon source, and industrial parameters, such as: lactic
acid yield (% of theoretical maximum from carbon source), volu-
metric productivity (g/L/h), and titer (g/L) [16,167].
The genomes of many strains of industrial importance have been
sequenced, which allows new developments in engineering lac-
tic acid bacteria for many industrial and commercial applications.
These studies allow us to better understand the metabolic path-
ways used for each substrate, the production of by-products, and
even stress conditions. This field of work has developed quickly, Fig. 5. The traditional process of lactic acid purification: extraction by precipitation.
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 233

Table 4 olutionized the idea of genetic engineering [168]. CRISPR-based


Main techniques available for product separation processes [46,179].
technologies have opened new opportunities for the develop-
Separation Process Principle ment of next-generation food micro-organisms and probiotics with
Equilibrium-based processes enhanced functionalities and safety. A review of the possibilities
Absorption Uses of ion-exchange resins to absorb the using CRISPR-based technologies for lactic acid bacteria is pre-
interest product sented by Hidalgo-Cantabrana et al. [170].
Distillation Based on heat and volatility of the compounds Genetic engineering is already a fundamental tool in the devel-
involved in the process
opment of an efficient bioprocess to produce lactic acid, bringing
Liquid-liquid extraction Separates chemicals from one solution to
(solvent extraction) another based on the different solubility of the many benefits of fast accomplishment for the industrial sector.
solute in two solvents
Affinity-based separation
8. Lactic acid separation and purification – downstream
Adsorption Uses of ion-exchange resins to adsorb the
interest product processes
Ion exchange Uses of ion-exchange resins to adsorb the
interest product The lactic acid purification process currently used has several
Simulated moving-bed Mobile phase moves countercurrent to a
economic and ecological implications for the final product. In gen-
chromatography stationary phase
Membrane separation
eral, costs of recovery and purification of biorefinery products,
Electrodialysis Extract ions from a solution through including lactic acid, represent 20–50% of the operating costs of
ion-exchange membrane to another solution the process [46]. In the conventional lactic acid recovery process
based on electric potential difference (extraction method by precipitation), lactic acid is precipitated with
Filtration Particles or molecules are retained in a
calcium hydroxide to form calcium lactate. This compound is recov-
membrane depending on the pore size, flow
and pressure used ered by a filtration process. Finally, sulfuric acid is used for the
Solid-liquid separation release of lactic acid, which generates a large amount of CaSO4 [46].
Conventional filtration Particles are retained in a membrane The diluted lactic acid product is then purified sequentially using
depending on the pore size, flow and pressure
activated carbon, evaporation, and crystallization processes (Fig. 5).
used
Precipitation and The product of interest is concentrated by
The precipitation extraction method has some disadvantages, such
crystallization evaporation and then crystallized as high cost due to the many reagents employed, the need for many
Reaction-separation systems for process intensification filtration steps, and the generation of large quantities of calcium
Reaction-membrane Bioreactors-membrane sulphate sludge as solid waste, leading to environmental problems
separation systems
[171–173]. It is known that approximately one ton of calcium sul-
Extractive fermentation Product extraction in situ
Reactive distillation Distillation preceded by a chemical reaction. phate of reduced economic value is produced for each ton of pure
Used when the product is of low volatility lactic acid produced [1], and this process can reach up to 50% of the
Reactive absorption Product extraction in situ using of total lactic acid production costs [172–174].
ion-exchange resins To obtain purified lactic acid it is necessary to eliminate: resid-
ual ions, residual sugar, cells, and other broth components at the
end of the process. As lactic acid is non-volatile and has a high affin-
tem against foreign DNA, such as phage or plasmids. According to ity for water molecules, the distillation, as well as the evaporation
Stefanovic et al. [168], analysis using this methodology provides processes, are energetically inefficient.
evidence of previous phage/strain interaction. It opens possibilities In this sense, research in regards to alternative separation and
for enhancing phage resistance of industrial strains such as Lacto- purification processes for lactic acid produced by fermentation
bacillus sp. Inspired by the mechanism of action of Type II CRISPR with lower costs and less environmental impact are necessary to
systems, genome editing represents a novel approach that has rev- increase process competitiveness.

Table 5
Most recent downstream studies for lactic acid recovery.

Separation method LA feedstock LA final concentration Yield (%) LA final purity Reference
(g/L) (wt %)

Short path evaporation Synthetic - - 7.13 ± 0.80 [215]


Reactive distillation Synthetic 347.68 99.94 ± 2.17 - [177]
Hybrid Short Path Evaporation Fermentation - - 89.7 [216]
Micro and nanofiltrations, Fermentation 937 - 99.7 [68]
softening, mono and bipolar
electrodialysis, anion and cation
exchange chromatography, and
distillation
Amberlite resins FPA 53 and CR Fermentation - 90 - [217]
5550
Fermentation and separation Fermentation 183.4 97 - [218]
integration system
(microfiltration and resin
adsorption)
Three-stage membrane- integrated Fermentation 250 96 95 [219]
hybrid reactor system
Membrane-Integrated Separation Fermentation - 76 99.5 [220]
Processes
Vapor permeation-assisted Fermentation - 95 - [221]
esterification
Cross-flow nanofiltration Fermentation - - 85.6 [180]
Molecular Distillation Fermentation - 74.09 95.6 [222]

LA: lactic acid


234 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

There are many techniques available for product separation pro- starchy biomass vs chemical synthesis, Catal. Today 239 (2015) 11–16,
cesses, as can be seen in Table 4. Advantages and disadvantages of http://dx.doi.org/10.1016/j.cattod.2014.05.039.
[9] R.P. John, K.M. Nampoothiri, A. Pandey, Fermentative production of lactic
some separation processes for lactic acid recovery are presented by acid from biomass: an overview on process developments and future
Komesu et al. [175]. perspectives, Appl. Microbiol. Biotechnol. 74 (2007) 524–534, http://dx.doi.
Many studies have been carried out using different extrac- org/10.1007/s00253-006-0779-6.
[10] S.I. Mussatto, M. Fernandes, I.M. Mancilha, I.C. Roberto, Effects of medium
tion and purification techniques for lactic acid. A glance at supplementation and pH control on lactic acid production from brewer’s
such processes leads to the use of solvent/solvent extraction, spent grain, Biochem. Eng. J. 40 (2008) 437–444, http://dx.doi.org/10.1016/j.
ion-exchange/adsorption, vacuum distillation, direct distilla- bej.2008.01.013.
[11] Y. Wang, Y. Tashiro, K. Sonomoto, Fermentative production of lactic acid
tion, esterification and hydrolysis methods, membrane sepa-
from renewable materials: recent achievements, prospects, and limits, J.
ration/membrane bioreactor, electro-dialysis, reverse osmosis, Biosci. Bioeng. 119 (2015) 10–18, http://dx.doi.org/10.1016/j.jbiosc.2014.06.
nanofiltration and ultrafiltration, liquid surfactant membrane 003.
[12] S.R. Kadam, S.S. Patil, K.B. Bastawde, J.M. Khire, D.V. Gokhale, Strain
extraction, chromatographic methods, drying, hybrid short path
improvement of Lactobacillus delbrueckii NCIM 2365 for lactic acid
evaporation, among others [176–184]. The most recent down- production, Process Biochem. 41 (2006) 120–126, http://dx.doi.org/10.1016/
stream studies are summarized in Table 5. j.procbio.2005.06.007.
[13] J. Vijayakumar, R. Aravindan, T. Viruthagiri, Recent trends in the production,
purification and application of lactic acid, Chem. Biochem. Eng. Q. 22 (2008)
9. Conclusions 245–264.
[14] Y.-J. Wee, H.-W. Ryu, Lactic acid production by Lactobacillus sp. RKY2 in a
cell-recycle continuous fermentation using lignocellulosic hydrolyzates as
The attainment of pure lactic acid to be used in several applica- inexpensive raw materials, Bioresour. Technol. 100 (2009) 4262–4270,
tions in different industrial sectors is a very complex process. In the http://dx.doi.org/10.1016/j.biortech.2009.03.074.
near future, the outlook for the lactic acid market is of continued [15] K. Hetényi, Á. Németh, B. Sevella, Role of pH-regulation in lactic acid
fermentation: second steps in a process improvement, Chem. Eng. Process.
growth, achieving new applications and better prices. Nowadays, Process Intensif. 50 (2011) 293–299, http://dx.doi.org/10.1016/j.cep.2011.
investments in the sector are focused on using new technolo- 01.008.
gies and new substrates to improve the production process and [16] M.A. Abdel-Rahman, K. Sonomoto, Opportunities to overcome the current
limitations and challenges for efficient microbial production of optically
achieve better micro-organism performance. Genetic engineering pure lactic acid, J. Biotechnol. 236 (2016) 176–192, http://dx.doi.org/10.
tools are developing quickly, focusing on creating more resistant 1016/j.jbiotec.2016.08.008.
and productive micro-organisms. It promises to be one of the sec- [17] E.M. Albuquerque, L.E.P. Borges, M.A. Fraga, Lactic acid production from
aqueous-phase selective oxidation of hydroxyacetone, J. Mol. Catal. A Chem.
tors with more innovation in the near future. This review presents 400 (2015) 64–70, http://dx.doi.org/10.1016/j.molcata.2015.02.005.
the basic settings that should be considered throughout the lac- [18] P. Gaspar, A.L. Carvalho, S. Vinga, H. Santos, A.R. Neves, From physiology to
tic acid production process, the nutritional needs of the producing systems metabolic engineering for the production of biochemicals by lactic
acid bacteria, Biotechnol. Adv. 31 (2013) 764–788, http://dx.doi.org/10.
micro-organisms, the metabolic implications of the most common
1016/j.biotechadv.2013.03.011.
factors of the fermentation, and the downstream process, as well [19] W. Bomrungnok, K. Sonomoto, S. Pinitglang, A. Wongwicharn, Single step
as the most recent achievements of the sector. lactic acid production from cassava starch by Lactobacillus plantarum SW14
in conventional continuous and continuous with high cell density, APCBEE
Procedia 2 (2012) 97–103, http://dx.doi.org/10.1016/j.apcbee.2012.06.018.
Funding [20] N. Narayanan, P.K. Roychoudhury, A. Srivastava, L(+) lactic acid
fermentation and its product polymerization, Electron. J. Biotechnol. 7
(2004) 167–179, http://dx.doi.org/10.2225/vol7-issue2-fulltext-7.
This work was supported by Foundation for Research of the State [21] B.J. Naveena, M. Altaf, K. Bhadriah, G. Reddy, Selection of medium
of São Paulo—FAPESP, Process 2013/26290-5. components by Plackett-Burman design for production of L(+) lactic acid by
Lactobacillus amylophilus GV6 in SSF using wheat bran, Bioresour. Technol.
96 (2005) 485–490, http://dx.doi.org/10.1016/j.biortech.2004.05.020.
Acknowledgments [22] G. Reddy, M. Altaf, B.J. Naveena, M. Venkateshwar, E.V. Kumar, Amylolytic
bacterial lactic acid fermentation—a review, Biotechnol. Adv. 26 (2008)
22–34, http://dx.doi.org/10.1016/j.biotechadv.2007.07.004.
The authors thank Espaço da Escrita – Pró Reitoria de Pesquisa [23] Y. Ramot, M. Haim-Zada, A.J. Domb, A. Nyska, Biocompatibility and safety of
– Unicamp – for the language services provided. PLA and its copolymers, Adv. Drug Deliv. Rev. 107 (2016) 153–162, http://
dx.doi.org/10.1016/j.addr.2016.03.012.
[24] J.W. Lee, H.U. Kim, S. Choi, J. Yi, S.Y. Lee, Microbial production of building
References block chemicals and polymers, Curr. Opin. Biotechnol. 22 (2011) 758–767,
http://dx.doi.org/10.1016/j.copbio.2011.02.011.
[1] R. Datta, S. Tsai, P. Bonsignore, S. Moon, J.R. Frank, Technological and [25] C. Boswell, Bioplastics aren’t the stretch they once seemed, Chem. Mark.
economical potencial of poly(lactic acid) and lactic acid derivatives, FEMS Rep. 260 (2001) pFR15.
Microbiol. Rev. 16 (1995) 221–231. [26] B.K. Ahring, J.J. Traverso, N. Murali, K. Srinivas, Continuous fermentation of
[2] R.A. de Oliveira, R. Maciel Filho, C.E.V. Rossell, High lactic acid production clarified corn stover hydrolysate for the production of lactic acid at high
from molasses and hydrolysed sugarcane bagasse, Chem. Eng. Trans. 50 yield and productivity, Biochem. Eng. J. 109 (2016) 162–169, http://dx.doi.
(2016) 307–312, http://dx.doi.org/10.3303/CET1650052. org/10.1016/j.bej.2016.01.012.
[3] K. Hofvendahl, B. Hahn-Hägerdal, Factors affecting the fermentative lactic [27] Y. Zhang, P.V. Vadlani, A. Kumar, P.R. Hardwidge, R. Govind, T. Tanaka, A.
acid production from renewable resources 1, Enzyme Microb. Technol. 26 Kondo, Enhanced D-lactic acid production from renewable resources using
(2000) 87–107, http://dx.doi.org/10.1016/S0141-0229(99)00155-6. engineered Lactobacillus plantarum, Appl. Microbiol. Biotechnol. 100 (2016)
[4] S. Zhou, T.B. Causey, A. Hasona, K.T. Shanmugam, L.O. Ingram, Production of 279–288, http://dx.doi.org/10.1007/s00253-015-7016-0.
optically pure D-lactic acid in mineral salts medium by metabolically [28] R.A. Gross, Biodegradable polymers for the environment, Science 297 (2002)
engineered Escherichia coli W3110, Appl. Environ. Microbiol. 69 (2003) 803–807, http://dx.doi.org/10.1126/science.297.5582.803.
399–407, http://dx.doi.org/10.1128/AEM.69.1.399-407.2003. [29] S. Yang, S.A. Madbouly, J.A. Schrader, G. Srinivasan, D. Grewell, K.G. McCabe,
[5] T. Ghaffar, M. Irshad, Z. Anwar, T. Aqil, Z. Zulifqar, A. Tariq, M. Kamran, N. M.R. Kessler, W.R. Graves, Characterization and biodegradation behavior of
Ehsan, S. Mehmood, Recent trends in lactic acid biotechnology: a brief bio-based poly(lactic acid) and soy protein blends for sustainable
review on production to purification, J. Radiat. Res. Appl. Sci. 7 (2014) horticultural applications, Green Chem. 17 (2015) 380–393, http://dx.doi.
222–229, http://dx.doi.org/10.1016/j.jrras.2014.03.002. org/10.1039/C4GC01482K.
[6] J. Blomqvist, RIS metropolis Monte Carlo studies of poly(L-lactic), poly(L, [30] A.P. Djukić-Vuković, L.V. Mojović, B.M. Jokić, S.B. Nikolić, J.D. Pejin, Lactic
D-lactic) and polyglycolic acids, Polymer (Guildf.) 42 (2001) 3515–3521, acid production on liquid distillery stillage by Lactobacillus rhamnosus
http://dx.doi.org/10.1016/S0032-3861(00)00704-7. immobilized onto zeolite, Bioresour. Technol. 135 (2013) 454–458, http://
[7] H. Tsuji, Autocatalytic hydrolysis of amorphous-made polylactides: effects dx.doi.org/10.1016/j.biortech.2012.10.066.
of L-lactide content, tacticity, and enantiomeric polymer blending, Polymer [31] T. Tanaka, M. Hoshina, S. Tanabe, K. Sakai, S. Ohtsubo, M. Taniguchi,
(Guildf.) 43 (2002) 1789–1796, http://dx.doi.org/10.1016/S0032- Production of D-lactic acid from defatted rice bran by simultaneous
3861(01)00752-2. saccharification and fermentation, Bioresour. Technol. 97 (2006) 211–217,
[8] G. Juodeikiene, D. Vidmantiene, L. Basinskiene, D. Cernauskas, E. Bartkiene, http://dx.doi.org/10.1016/j.biortech.2005.02.025.
D. Cizeikiene, Green metrics for sustainability of biobased lactic acid from
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 235

[32] E. Castro-Aguirre, F. Iñiguez-Franco, H. Samsudin, X. Fang, R. Auras, [55] F. Cherubini, The biorefinery concept: using biomass instead of oil for
Poly(lactic acid)—mass production, processing, industrial applications, and producing energy and chemicals, Energy Convers. Manage. 51 (2010)
end of life, Adv. Drug Deliv. Rev. 107 (2016) 333–366, http://dx.doi.org/10. 1412–1421, http://dx.doi.org/10.1016/j.enconman.2010.01.015.
1016/j.addr.2016.03.010. [56] J. Sikder, S. Chakraborty, V. Sharma, E. Drioli, Kinetic of lactic acid production
[33] M.A. Abdel-Rahman, Y. Tashiro, K. Sonomoto, Lactic acid production from from sugarcane juice using Lactobacillus plantarum NCIM 2912, Asia-Pacific
lignocellulose-derived sugars using lactic acid bacteria: overview and limits, J. Chem. Eng. 9 (2014) 374–381, http://dx.doi.org/10.1002/apj.1806.
J. Biotechnol. 156 (2011) 286–301, http://dx.doi.org/10.1016/j.jbiotec.2011. [57] P. Laopaiboon, A. Thani, V. Leelavatcharamas, L. Laopaiboon, Acid hydrolysis
06.017. of sugarcane bagasse for lactic acid production, Bioresour. Technol. 101
[34] S. Hammouche, D. Hammouche, M. McNicholas, Biodegradable bone (2010) 1036–1043, http://dx.doi.org/10.1016/j.biortech.2009.08.091.
regeneration synthetic scaffolds: in tissue engineering, Curr. Stem Cell Res. [58] Y. Wang, H. Meng, D. Cai, B. Wang, P. Qin, Z. Wang, T. Tan, Improvement of
Ther. 7 (2012) 134–142, http://dx.doi.org/10.2174/157488812799219018. L-lactic acid productivity from sweet sorghum juice by repeated batch
[35] R.P.F. Lanao, A.M. Jonker, J.G.C. Wolke, J. a Jansen, J.C.M. van Hest, S.C.G. fermentation coupled with membrane separation, Bioresour. Technol. 211
Leeuwenburgh, Physicochemical properties and applications of (2016) 291–297, http://dx.doi.org/10.1016/j.biortech.2016.03.095.
poly(lactic-co-glycolic acid) for use in bone regeneration, Tissue Eng. Part B [59] Y. Wang, C. Chen, D. Cai, Z. Wang, P. Qin, T. Tan, The optimization of L-lactic
Rev. 19 (2013) 380–390, http://dx.doi.org/10.1089/ten.teb.2012.0443. acid production from sweet sorghum juice by mixed fermentation of
[36] M.M. Sakata, M.C. Alberto-Rincon, E.A.R. Duek, Estudo da interação Bacillus coagulans and Lactobacillus rhamnosus under unsterile conditions,
polímero/cartilagem/osso utilizando poli (ácido lático-co-ácido glicólico) e Bioresour. Technol. 218 (2016) 1098–1105, http://dx.doi.org/10.1016/j.
poli (p-dioxanona) em condilo femural de coelhos, Polímeros 14 (2004) biortech.2016.07.069.
176–180, http://dx.doi.org/10.1590/S0104-14282004000300013. [60] M. Kuisma, H. Kahiluoto, J. Havukainen, E. Lehtonen, M. Luoranen, T.
[37] M.J. Biddy, C. Scarlata, C. Kinchin, Chemicals from Biomass: A Market Myllymaa, J. Grönroos, M. Horttanainen, Understanding biorefining
Assessment of Bioproducts with Near-Term Potential, Golden, CO (United efficiency—the case of agrifood waste, Bioresour. Technol. 135 (2013)
States), 2016, http://dx.doi.org/10.2172/1244312. 588–597, http://dx.doi.org/10.1016/j.biortech.2012.11.038.
[38] T.H. Kwan, D. Pleissner, K.Y. Lau, J. Venus, A. Pommeret, C.S.K. Lin, [61] G. Dedenaro, S. Costa, I. Rugiero, P. Pedrini, E. Tamburini, Valorization of
Techno-economic analysis of a food waste valorization process via agri-food waste via fermentation: production of L-lactic acid as a building
microalgae cultivation and co-production of plasticizer, lactic acid and block for the synthesis of biopolymers, Appl. Sci. 6 (2016) 379, http://dx.doi.
animal feed from algal biomass and food waste, Bioresour. Technol. 198 org/10.3390/app6120379.
(2015) 292–299, http://dx.doi.org/10.1016/j.biortech.2015.09.003. [62] A. Djukić-Vuković, D. Mladenović, M. Radosavljević, S. Kocić-Tanackov, J.
[39] S. Farzad, M.A. Mandegari, M. Guo, K.F. Haigh, N. Shah, J.F. Görgens, Pejin, L. Mojović, Wastes from bioethanol and beer productions as substrates
Multi-product biorefineries from lignocelluloses: a pathway to revitalisation for L(+) lactic acid production—a comparative study, Waste Manage. 48
of the sugar industry? Biofuels 10 (2017) 87, http://dx.doi.org/10.1186/ (2016) 478–482, http://dx.doi.org/10.1016/j.wasman.2015.11.031.
s13068-017-0761-9. [63] M. Probst, J. Walde, T. Pümpel, A.O. Wagner, H. Insam, A closed loop for
[40] J. Sikder, M. Roy, P. Dey, P. Pal, Techno-economic analysis of a municipal organic solid waste by lactic acid fermentation, Bioresour.
membrane-integrated bioreactor system for production of lactic acid from Technol. 175 (2015) 142–151, http://dx.doi.org/10.1016/j.biortech.2014.10.
sugarcane juice, Biochem. Eng. J. 63 (2012) 81–87, http://dx.doi.org/10. 034.
1016/j.bej.2011.11.004. [64] T.H. Kwan, Y. Hu, C.S.K. Lin, Valorisation of food waste via fungal hydrolysis
[41] G. Liu, J. Sun, J. Zhang, Y. Tu, J. Bao, High titer L-lactic acid production from and lactic acid fermentation with Lactobacillus casei Shirota, Bioresour.
corn stover with minimum wastewater generation and techno-economic Technol. 217 (2016) 129–136, http://dx.doi.org/10.1016/j.biortech.2016.01.
evaluation based on Aspen plus modeling, Bioresour. Technol. 198 (2015) 134.
803–810, http://dx.doi.org/10.1016/j.biortech.2015.09.098. [65] Y. Wu, H. Ma, M. Zheng, K. Wang, Lactic acid production from acidogenic
[42] A. Gezae Daful, J.F. Görgens, Techno-economic analysis and environmental fermentation of fruit and vegetable wastes, Bioresour. Technol. 191 (2015)
impact assessment of lignocellulosic lactic acid production, Chem. Eng. Sci. 53–58, http://dx.doi.org/10.1016/j.biortech.2015.04.100.
162 (2017) 53–65, http://dx.doi.org/10.1016/j.ces.2016.12.054. [66] T. Overbeck, J.L. Steele, J.R. Broadbent, Fermentation of de-oiled algal
[43] S.I. Mussatto, J. Moncada, I.C. Roberto, C.A. Cardona, Techno-economic biomass by Lactobacillus casei for production of lactic acid, Bioprocess
analysis for brewer’s spent grains use on a biorefinery concept: the Brazilian Biosyst. Eng. 39 (2016) 1817–1823, http://dx.doi.org/10.1007/s00449-016-
case, Bioresour. Technol. 148 (2013) 302–310, http://dx.doi.org/10.1016/j. 1656-z.
biortech.2013.08.046. [67] Ö. Tirpanalan, M. Reisinger, M. Smerilli, F. Huber, M. Neureiter, W. Kneifel, S.
[44] M.I. González, S. Álvarez, F. Riera, R. Álvarez, Economic evaluation of an Novalin, Wheat bran biorefinery—an insight into the process chain for the
integrated process for lactic acid production from ultrafiltered whey, J. Food production of lactic acid, Bioresour. Technol. 180 (2015) 242–249, http://dx.
Eng. 80 (2007) 553–561, http://dx.doi.org/10.1016/j.jfoodeng.2006.06.021. doi.org/10.1016/j.biortech.2015.01.021.
[45] C. Åkerberg, G. Zacchi, An economic evaluation of the fermentative [68] D. Pleissner, A.-K. Neu, K. Mehlmann, R. Schneider, G.I. Puerta-Quintero, J.
production of lactic acid from wheat flour, Bioresour. Technol. 75 (2000) Venus, Fermentative lactic acid production from coffee pulp hydrolysate
119–126, http://dx.doi.org/10.1016/S0960-8524(00)00057-2. using Bacillus coagulans at laboratory and pilot scales, Bioresour. Technol.
[46] S. Ramaswamy, H.J. Huang, B.V. Ramarao, Separation and Purification 218 (2016) 167–173, http://dx.doi.org/10.1016/j.biortech.2016.06.078.
Technologies in Biorefineries, John Wiley & Sons, Ltd, Chichester, UK, 2013, [69] M.A. Mandegari, S. Farzad, E. van Rensburg, J.F. Görgens, Multi-criteria
http://dx.doi.org/10.1002/9781118493441. analysis of a biorefinery for co-production of lactic acid and ethanol from
[47] R. Kumar, H. Nanavati, S.B. Noronha, S.M. Mahajani, A continuous process sugarcane lignocellulose, Biofuels Bioprod. Biorefin. 11 (2017) 971–990,
for the recovery of lactic acid by reactive distillation, J. Chem. Technol. http://dx.doi.org/10.1002/bbb.1801.
Biotechnol. 81 (2006) 1767–1777, http://dx.doi.org/10.1002/jctb.1603. [70] R. Parajuli, M.T. Knudsen, M. Birkved, S.N. Djomo, A. Corona, T. Dalgaard,
[48] C.-Y. Su, C.-C. Yu, I.-L. Chien, J.D. Ward, Plant-wide economic comparison of Environmental impacts of producing bioethanol and biobased lactic acid
lactic acid recovery processes by reactive distillation with different alcohols, from standalone and integrated biorefineries using a consequential and an
Ind. Eng. Chem. Res. 52 (2013) 11070–11083, http://dx.doi.org/10.1021/ attributional life cycle assessment approach, Sci. Total Environ. 598 (2017)
ie303192x. 497–512, http://dx.doi.org/10.1016/j.scitotenv.2017.04.087.
[49] B.H. Lunelli, R.R. Andrade, D.I.P. Atala, M.R. Wolf Maciel, F. Maugeri Filho, R. [71] G. Finnveden, M.Z. Hauschild, T. Ekvall, J. Guinée, R. Heijungs, S. Hellweg, A.
Maciel Filho, Production of lactic acid from sucrose: strain selection, Koehler, D. Pennington, S. Suh, Recent developments in life cycle
fermentation, and kinetic modeling, Appl. Biochem. Biotechnol. 161 (2010) assessment, J. Environ. Manage. 91 (2009) 1–21, http://dx.doi.org/10.1016/j.
227–237, http://dx.doi.org/10.1007/s12010-009-8828-0. jenvman.2009.06.018.
[50] A.J.R.R. Lasprilla, G.A.R.R. Martinez, B.H. Lunelli, A.L. Jardini, R.M. Filho, [72] J.B. Guineı́e, R. Heijungs, G. Huppes, A. Zamagni, P. Masoni, R. Buonamici, T.
Poly-lactic acid synthesis for application in biomedical devices—a review, Ekvall, T. Rydberg, Life cycle assessment: past, present, and future, Environ.
Biotechnol. Adv. 30 (2012) 321–328, http://dx.doi.org/10.1016/j.biotechadv. Sci. Technol. 45 (2011) 90–96, http://dx.doi.org/10.1021/es101316v.
2011.06.019. [73] M.F. Cosate de Andrade, P.M.S. Souza, O. Cavalett, A.R. Morales, Life cycle
[51] L.V. Reddy, Y.-M. Kim, J.-S. Yun, H.-W. Ryu, Y.-J. Wee, L-Lactic acid assessment of poly(lactic acid) (PLA): comparison between chemical
production by combined utilization of agricultural bioresources as recycling, mechanical recycling and composting, J. Polym. Environ. 24
renewable and economical substrates through batch and repeated-batch (2016) 372–384, http://dx.doi.org/10.1007/s10924-016-0787-2.
fermentation of Enterococcus faecalis RKY1, Bioresour. Technol. 209 (2016) [74] J.-M. Park, A. Kondo, J.-S. Chang, C. Perry Chou, P. Monsan, Biorefineries,
187–194, http://dx.doi.org/10.1016/j.biortech.2016.02.115. Bioresour. Technol. 135 (2013) 1, http://dx.doi.org/10.1016/j.biortech.2013.
[52] M. Palmeros Parada, P. Osseweijer, J.A. Posada Duque, Sustainable 03.132.
biorefineries, an analysis of practices for incorporating sustainability in [75] F.K. Adom, J.B. Dunn, Life cycle analysis of corn-stover-derived
biorefinery design, Ind. Crops Prod. 106 (2017) 105–123, http://dx.doi.org/ polymer-grade L-lactic acid and ethyl lactate: greenhouse gas emissions and
10.1016/j.indcrop.2016.08.052. fossil energy consumption, Biofuels Bioprod. Biorefin. 11 (2017) 258–268,
[53] J.B. Moncada, V.M. Aristizábal, C.A. Cardona, Design strategies for http://dx.doi.org/10.1002/bbb.1734.
sustainable biorefineries, Biochem. Eng. J. 116 (2016) 122–134, http://dx. [76] P.R. Gruber, Commodity polymers from renewable resources: polylactic
doi.org/10.1016/j.bej.2016.06.009. acid, in: Carbon Manag. Implic. R D Chem. Sci. Technol., National Academy of
[54] M. Sillanpää, C. Ncibi, A Sustainable Bioeconomy, Springer International Sciences, Washington, D.C, 2001, p. 223.
Publishing, Cham, 2017, http://dx.doi.org/10.1007/978-3-319-55637-6. [77] G.S. Lorite, J.M.R. Rocha, N. Miilumäki, P. Saavalainen, T. Selkälä, G.
Morales-Cid, M.P. Gonçalves, E. Pongrácz, C.M.R. Rocha, G. Toth, Evaluation
of physicochemical/microbial properties and life cycle assessment (LCA) of
236 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

PLA-based nanocomposite active packaging, LWT—Food Sci. Technol. 75 [101] H.-Y. Choi, H.-K. Ryu, K.-M. Park, E.G. Lee, H. Lee, S.-W. Kim, E.-S. Choi, Direct
(2017) 305–315, http://dx.doi.org/10.1016/j.lwt.2016.09.004. lactic acid fermentation of Jerusalem artichoke tuber extract using
[78] W.J. Groot, T. Borén, Life cycle assessment of the manufacture of lactide and Lactobacillus paracasei without acidic or enzymatic inulin hydrolysis,
PLA biopolymers from sugarcane in Thailand, Int. J. Life Cycle Assess. 15 Bioresour. Technol. 114 (2012) 745–747, http://dx.doi.org/10.1016/j.
(2010) 970–984, http://dx.doi.org/10.1007/s11367-010-0225-y. biortech.2012.03.075.
®
[79] E.T.H. Vink, S. Davies, J.J. Kolstad, The eco-profile for current Ingeo [102] A.H. Jawad, A.F.M.M. Alkarkhi, O.C. Jason, A.M. Easa, N.A. Nik Norulaini,
polylactide production, Ind. Biotechnol. 6 (2010) 212–224, http://dx.doi.org/ N.A.N. Norulaini, Production of the lactic acid from mango peel
10.1089/ind.2010.6.212. waste—factorial experiment, J. King Saud Univ. Sci. 25 (2013) 39–45, http://
[80] E.T.H. Vink, K.R. Rábago, D.A. Glassner, P.R. Gruber, Applications of life cycle dx.doi.org/10.1016/j.jksus.2012.04.001.
assessment to NatureWorksTM polylactide (PLA) production, Polym. Degrad. [103] A. Neu, D. Pleissner, K. Mehlmann, R. Schneider, G.I. Puerta-quintero, J.
Stab. 80 (2003) 403–419, http://dx.doi.org/10.1016/S0141-3910(02)00372- Venus, Fermentative utilization of coffee mucilage using Bacillus coagulans
5. and investigation of down-stream processing of fermentation broth for
[81] E.T.H. Vink, S. Davies, Life cycle inventory and impact assessment datafor optically pure L(+)-lactic acid production, Bioresour. Technol. 211 (2016)
2014 IngeoTM polylactide production, Ind. Biotechnol. 11 (2015) 167–180, 398–405, http://dx.doi.org/10.1016/j.biortech.2016.03.122.
http://dx.doi.org/10.1089/ind.2015.0003. [104] W. Fu, A.P. Mathews, Lactic acid production from lactose by Lactobacillus
[82] T.A. Hottle, M.M. Bilec, A.E. Landis, Biopolymer production and end of life plantarum: kinetic model and effects of pH, substrate, and oxygen, Biochem.
comparisons using life cycle assessment, Resour. Conserv. Recycl. 122 Eng. J. 3 (1999) 163–170, http://dx.doi.org/10.1016/S1369-703X(99)00014-
(2017) 295–306, http://dx.doi.org/10.1016/j.resconrec.2017.03.002. 5.
[83] W. Schimidell, U.A. Lima, E. Aquarone, W. Borzani, Biotecnologia Industrial: [105] U. Farooq, F.M. Anjum, T. Zahoor, Sajjad-Ur-Rahman, M.A. Randhawa, A.
Engenharia química, Edgard Blucher LTDA, São Paulo, 2001. Ahmed, K. Akram, Optimization of lactic acid production from cheap raw
[84] J. Ouyang, R. Ma, Z. Zheng, C. Cai, M. Zhang, T. Jiang, Open fermentative material: sugarcane molasses, Pak. J. Bot. 44 (2012) 333–3338.
production of L-lactic acid by Bacillus sp. strain NL01 using lignocellulosic [106] A. Komesu, J.A.R. De Oliveira, L.H. da, S. Martins, M.R. Wolf Maciel, R. Maciel
hydrolyzates as low-cost raw material, Bioresour. Technol. 135 (2013) Filho, Lactic acid production to purification: a review, BioResources 12
475–480, http://dx.doi.org/10.1016/j.biortech.2012.09.096. (2017) 4364–4383.
[85] T. Gao, Y. Wong, C. Ng, K. Ho, L-lactic acid production by Bacillus subtilis [107] S. Tosungnoen, K. Chookietwattana, S. Dararat, Lactic acid production from
MUR1, Bioresour. Technol. 121 (2012) 105–110, http://dx.doi.org/10.1016/j. repeated-batch and simultaneous saccharification and fermentation of
biortech.2012.06.108. cassava starch wastewater by amylolytic Lactobacillus Plantarum MSUL 702,
[86] F.A. Castillo Martinez, E.M. Balciunas, J.M. Salgado, J.M. Domínguez APCBEE Procedia 8 (2014) 204–209, http://dx.doi.org/10.1016/j.apcbee.
González, A. Converti, R.P.D.S. Oliveira, Lactic acid properties, applications 2014.03.028.
and production: a review, Trends Food Sci. Technol. 30 (2013) 70–83, http:// [108] D.J. Tenenbaum, Food vs. Fuel: diversion of crops could cause more hunger,
dx.doi.org/10.1016/j.tifs.2012.11.007. Environ. Heal. Perspect. Credit. 116 (2008) 254–257.
[87] M.A. Abdel-Rahman, Y. Tashiro, K. Sonomoto, Recent advances in lactic acid [109] S.I. Musatto, J.A. Teixeira, Lignocellulose as raw material in fermentation
production by microbial fermentation processes, Biotechnol. Adv. 31 (2013) processes, Commun. Curr. Res. Technol. Educ. Top. Appl. Microbiol. Microb.
877–902, http://dx.doi.org/10.1016/j.biotechadv.2013.04.002. Biotechnol. 2 (2010) 897–907.
[88] Z. Lu, F. He, Y. Shi, M. Lu, L. Yu, Fermentative production of L(+)-lactic acid [110] E. Palmqvist, B. Hahn-Hagerdal, Fermentation of lignocellulosic
using hydrolyzed acorn starch, persimmon juice and wheat bran hydrolysates. II: inhibitors and mechanisms of inhibition, Bioresour.
hydrolysate as nutrients, Bioresour. Technol. 101 (2010) 3642–3648, http:// Technol. 74 (2000) 25–33.
dx.doi.org/10.1016/j.biortech.2009.12.119. [111] E.C. van der Pol, E. Vaessen, R.A. Weusthuis, G. Eggink, Identifying inhibitory
[89] C.M. Nguyen, G.J. Choi, Y.H. Choi, K.S. Jang, J.-C. Kim, D- and L-lactic acid effects of lignocellulosic by-products on growth of lactic acid producing
production from fresh sweet potato through simultaneous saccharification micro-organisms using a rapid small-scale screening method, Bioresour.
and fermentation, Biochem. Eng. J. 81 (2013) 40–46, http://dx.doi.org/10. Technol. 209 (2016) 297–304, http://dx.doi.org/10.1016/j.biortech.2016.03.
1016/j.bej.2013.10.003. 037.
[90] M. Bishai, S. De, B. Adhikari, R. Banerjee, Zizyphus oenophlia: a potent [112] M. Alkasrawi, A. Rudolf, G. Lidén, G. Zacchi, Influence of strain and
substrate for lactic acid production, Bioresour. Technol. 133 (2013) cultivation procedure on the performance of simultaneous saccharification
627–629, http://dx.doi.org/10.1016/j.biortech.2012.12.049. and fermentation of steam pretreated spruce, Enzyme Microb. Technol. 38
[91] Z. Li, J. Lu, Z. Yang, L. Han, T. Tan, Utilization of white rice bran for production (2006) 279–286, http://dx.doi.org/10.1016/j.enzmictec.2005.08.024.
of L-lactic acid, Biomass Bioenergy 39 (2012) 53–58, http://dx.doi.org/10. [113] L. Zhang, X. Li, Q. Yong, S. Yang, J. Ouyang, S. Yu, Impacts of
1016/j.biombioe.2011.12.039. lignocellulose-derived inhibitors on L-lactic acid fermentation by Rhizopus
[92] M. Watanabe, M. Makino, N. Kaku, M. Koyama, K. Nakamura, K. Sasano, oryzae, Bioresour. Technol. 203 (2016) 173–180, http://dx.doi.org/10.1016/j.
Fermentative L-(+)-lactic acid production from non-sterilized rice washing biortech.2015.12.014.
drainage containing rice bran by a newly isolated lactic acid bacteria [114] L.J. Jönsson, C. Martín, Pretreatment of lignocellulose: formation of
without any additions of nutrients, J. Biosci. Bioeng. 115 (2013) 449–452, inhibitory by-products and strategies for minimizing their effects,
http://dx.doi.org/10.1016/j.jbiosc.2012.11.001. Bioresour. Technol. 199 (2016) 103–112, http://dx.doi.org/10.1016/j.
[93] K. Zhao, Q. Qiao, D. Chu, H. Gu, T.H. Dao, J. Zhang, J. Bao, Simultaneous biortech.2015.10.009.
saccharification and high titer lactic acid fermentation of corn stover using a [115] V. Juturu, J.C. Wu, Microbial production of lactic acid: the latest
newly isolated lactic acid bacterium Pediococcus acidilactici DQ2, Bioresour. development, Crit. Rev. Biotechnol. 36 (2016) 967–977, http://dx.doi.org/10.
Technol. 135 (2013) 481–489, http://dx.doi.org/10.1016/j.biortech.2012.09. 3109/07388551.2015.1066305.
063. [116] C.M. Nguyen, J.-S. Kim, J.K. Song, G.J. Choi, Y.H. Choi, K.S. Jang, J.-C. Kim,
[94] F. Cui, Y. Li, C. Wan, Lactic acid production from corn stover using mixed D-Lactic acid production from dry biomass of Hydrodictyon reticulatum by
cultures of Lactobacillus rhamnosus and Lactobacillus brevis, Bioresour. simultaneous saccharification and co-fermentation using Lactobacillus
Technol. 102 (2011) 1831–1836, http://dx.doi.org/10.1016/j.biortech.2010. coryniformis subsp, torquens, Biotechnol. Lett. 34 (2012) 2235–2240, http://
09.063. dx.doi.org/10.1007/s10529-012-1023-3.
[95] M.G. Adsul, A.J. Varma, D.V. Gokhale, Lactic acid production from waste [117] S. Hirayama, R. Ueda, Production of optically pure D-lactic acid by
sugarcane bagasse derived cellulose, Green Chem. 9 (2007) 58–62, http://dx. Nannochlorum sp. 26A4, Appl. Biochem. Biotechnol. 119 (2004) 71–77.
doi.org/10.1039/B605839F. [118] B. Zhang, P. He, N. Ye, L. Shao, Enhanced isomer purity of lactic acid from the
[96] Z. Lu, M. Lu, F. He, L. Yu, An economical approach for D-lactic acid non-sterile fermentation of kitchen wastes, Bioresour. Technol. 99 (2008)
production utilizing unpolished rice from aging paddy as major nutrient 855–862, http://dx.doi.org/10.1016/j.biortech.2007.01.010.
source, Bioresour. Technol. 100 (2009) 2026–2031, http://dx.doi.org/10. [119] D. Pleissner, W.C. Lam, Z. Sun, C.S.K. Lin, Food waste as nutrient source in
1016/j.biortech.2008.10.015. heterotrophic microalgae cultivation, Bioresour. Technol. 137 (2013)
[97] C.M. Nguyen, J.-S. Kim, T.N. Nguyen, S.K. Kim, G.J. Choi, Y.H. Choi, K.S. Jang, 139–146, http://dx.doi.org/10.1016/j.biortech.2013.03.088.
J.-C. Kim, Production of L- and D-lactic acid from waste Curcuma longa [120] V. Kitpreechavanich, A. Hayami, A. Talek, C.F.S. Chin, Y. Tashiro, K. Sakai,
biomass through simultaneous saccharification and cofermentation, Simultaneous production of L-lactic acid with high optical activity and a soil
Bioresour. Technol. 146 (2013) 35–43, http://dx.doi.org/10.1016/j.biortech. amendment with food waste that demonstrates plant growth promoting
2013.07.035. activity, J. Biosci. Bioeng. 122 (2016) 105–110, http://dx.doi.org/10.1016/j.
[98] N.K. Budhavaram, Z. Fan, Production of lactic acid from paper sludge using jbiosc.2015.12.017.
acid-tolerant, thermophilic Bacillus coagulan strains, Bioresour. Technol. 100 [121] Y. Tashiro, S. Inokuchi, P. Poudel, Y. Okugawa, H. Miyamoto, H. Miayamoto,
(2009) 5966–5972, http://dx.doi.org/10.1016/j.biortech.2009.01.080. K. Sakai, Novel pH control strategy for efficient production of optically active
[99] C.M. Nguyen, J.-S. Kim, H.J. Hwang, M.S. Park, G.J. Choi, Y.H. Choi, K.S. Jang, L-lactic acid from kitchen refuse using a mixed culture system, Bioresour.
J.-C. Kim, Production of L-lactic acid from a green microalga, Hydrodictyon Technol. 216 (2016) 52–59, http://dx.doi.org/10.1016/j.biortech.2016.05.
reticulum, by Lactobacillus paracasei LA104 isolated from the traditional 031.
Korean food, makgeolli, Bioresour. Technol. 110 (2012) 552–559, http://dx. [122] J. Tang, X.C. Wang, Y. Hu, H.H. Ngo, Y. Li, Dynamic membrane-assisted
doi.org/10.1016/j.biortech.2012.01.079. fermentation of food wastes for enhancing lactic acid production, Bioresour.
[100] L. Ye, X. Zhou, M.S. Bin Hudari, Z. Li, J.C. Wu, Highly efficient production of Technol. 234 (2017) 40–47, http://dx.doi.org/10.1016/j.biortech.2017.03.
L-lactic acid from xylose by newly isolated Bacillus coagulans C106, 019.
Bioresour. Technol. 132 (2013) 38–44, http://dx.doi.org/10.1016/j.biortech. [123] J. Tang, X.C. Wang, Y. Hu, Y. Zhang, Y. Li, Effect of pH on lactic acid
2013.01.011. production from acidogenic fermentation of food waste with different types
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 237

of inocula, Bioresour. Technol. 224 (2017) 544–552, http://dx.doi.org/10. sakei and other lactic acid bacteria, J. Gen. Appl. Microbiol. 47 (2001)
1016/j.biortech.2016.11.111. 223–239, http://dx.doi.org/10.2323/jgam.47.223.
[124] N. Murakami, M. Oba, M. Iwamoto, Y. Tashiro, T. Noguchi, K. Bonkohara, [148] J. Stiles, S. Penkar, M. Plocková, J. Chumchalová, L.B. Bullerman, Antifungal
M.A. Abdel-Rahman, T. Zendo, M. Shimoda, K. Sakai, K. Sonomoto, L-Lactic activity of sodium acetate and Lactobacillus rhamnosus, J. Food Prot. 65
acid production from glycerol coupled with acetic acid metabolism by (2002) 1188–1191, http://dx.doi.org/10.1016/j.ijfoodmicro.2013.12.016.
Enterococcus faecalis without carbon loss, J. Biosci. Bioeng. 121 (2016) 89–95, [149] K. Kyla-Nikkila, M. Hujanen, M. Leisola, A. Palva, Metabolic engineering of
http://dx.doi.org/10.1016/j.jbiosc.2015.05.009. Lactobacillus helveticus CNRZ32 for production of pure L-(+)-lactic acid, Appl.
[125] A.-A. Hong, K.-K. Cheng, F. Peng, S. Zhou, Y. Sun, C.-M. Liu, D.-H. Liu, Strain Environ. Microbiol. 66 (2000) 3835–3841, http://dx.doi.org/10.1128/AEM.
isolation and optimization of process parameters for bioconversion of 66.9.3835-3841.2000.
glycerol to lactic acid, J. Chem. Technol. Biotechnol. 84 (2009) 1576–1581, [150] D.D. Axe, J.E. Bailey, Transport of lactate and acetate through the energized
http://dx.doi.org/10.1002/jctb.2209. cytoplasmic membrane of Escherichia coli, Biotechnol. Bioeng. 47 (1995)
[126] Y. Doi, Y. Ikegami, Pyruvate formate-lyase is essential for 8–19.
fumarate-independent anaerobic glycerol utilization in the Enterococcus [151] S. Nakano, C.U. Ugwu, Y. Tokiwa, Efficient production of D-(−)-lactic acid
faecalis strain W11, J. Bacteriol. 196 (2014) 2472–2480, http://dx.doi.org/10. from broken rice by Lactobacillus delbrueckii using Ca(OH)2 as a neutralizing
1128/JB.01512-14. agent, Bioresour. Technol. 104 (2012) 791–794, http://dx.doi.org/10.1016/j.
[127] S. Bulut, M. Elibol, D. Ozer, Effect of different carbon sources on L(+) −lactic biortech.2011.10.017.
acid production by Rhizopus oryzae, Biochem. Eng. J. 21 (2004) 33–37, http:// [152] P.D. Cotter, C. Hill, Surviving the acid test: responses of gram-positive
dx.doi.org/10.1016/j.bej.2004.04.006. bacteria to low pH, Microbiol. Mol. Biol. Rev. 67 (2003) 429–453, http://dx.
[128] G. Chotani, T. Dodge, A. Hsu, M. Kumar, R. LaDuca, D. Trimbur, W. Weyler, K. doi.org/10.1128/MMBR.67.3.429-453.2003.
Sanford, The commercial production of chemicals using pathway [153] Z. Zhai, F.P. Douillard, H. An, G. Wang, X. Guo, Y. Luo, Y. Hao, Proteomic
engineering, Biochim. Biophys. Acta—Protein Struct. Mol. Enzymol. 2000 characterization of the acid tolerance response in Lactobacillus delbrueckii
(1543) 434–455, http://dx.doi.org/10.1016/S0167-4838(00)00234-X. subsp. bulgaricus CAUH1 and functional identification of a novel acid
[129] Committee on Biobased Industrial Products (Ed.), Biobased Industrial stress-related transcriptional regulator Ldb0677, Environ. Microbiol. 16
Products, National Academies Press, Washington, D.C, 1722, http://dx.doi. (2014) 1524–1537, http://dx.doi.org/10.1111/1462-2920.12280.
org/10.17226/5295. [154] E.M. Lim, S.D. Ehrlich, E. Maguin, Identification of stress-inducible proteins
[130] L. Ye, H. Zhao, Z. Li, J.C. Wu, Improved acid tolerance of Lactobacillus pentosus in Lactobacillus delbrueckii subsp. bulgaricus, Electrophoresis 21 (2000)
by error-prone whole genome amplification, Bioresour. Technol. 135 (2013) 2557–2561, http://dx.doi.org/10.1002/1522-
459–463, http://dx.doi.org/10.1016/j.biortech.2012.10.042. 2683(20000701)21:12<2557:AID-ELPS2557>3.0.CO;2-B.
[131] S. Rodríguez Couto, Exploitation of biological wastes for the production of [155] S. Siragusa, M. De Angelis, M. Calasso, D. Campanella, F. Minervini, R. Di
value-added products under solid-state fermentation conditions, Cagno, M. Gobbetti, M. De Angelis, M. Calasso, D. Campanella, F. Minervini,
Biotechnol. J. 3 (2008) 859–870, http://dx.doi.org/10.1002/biot.200800031. R. Di Cagno, M. Gobbetti, Fermentation and proteome profiles of
[132] S. Salminen, A. Von Wright, A. Ouwehand, Lactic Acid Bacteria: Lactobacillus plantarum strains during growth under food-like conditions, J.
Microbiological and Functional Aspects, 3rd ed., Marcel Dekker, Inc, New Proteom. 96 (2014) 366–380, http://dx.doi.org/10.1016/j.jprot.2013.11.003.
York, 2004. [156] J.R. Broadbent, R.L. Larsen, V. Deibel, J.L. Steele, Physiological and
[133] E. Caplice, Food fermentations: role of microorganisms in food production transcriptional response of Lactobacillus casei ATCC 334 to acid stress, J.
and preservation, Int. J. Food Microbiol. 50 (1999) 131–149, http://dx.doi. Bacteriol. 192 (2010) 2445–2458, http://dx.doi.org/10.1128/JB.01618-09.
org/10.1016/S0168-1605(99)00082-3. [157] J. Koponen, K. Laakso, K. Koskenniemi, M. Kankainen, K. Savijoki, T.A.
[134] V. Arasaratnam, A. Senthuran, K. Balasubramaniam, Supplementation of Nyman, W.M. de Vos, S. Tynkkynen, N. Kalkkinen, P. Varmanen, Effect of
whey with glucose and different nitrogen sources for lactic acid production acid stress on protein expression and phosphorylation in Lactobacillus
by Lactictobacillus delbrueckii, Enzyme Microb. Technol. 19 (1996) 482–486. rhamnosus GG, J. Proteom. 75 (2012) 1357–1374, http://dx.doi.org/10.1016/
[135] G.C. Cox, R.D. MacBean, Lactic acid production by Lactobacillus bulgaricus in j.jprot.2011.11.009.
supplemented whey ultrafiltrate, Aust. J. Dairy Technol. 32 (1977) 19–22. [158] Y.-M. Zhang, C.O. Rock, Membrane lipid homeostasis in bacteria, Nat. Rev.
[136] A. Nancib, N. Nancib, D. Meziane-Cherif, A. Boubendir, M. Fick, J. Boudrant, Microbiol. 6 (2008) 222–233, http://dx.doi.org/10.1038/nrmicro1839.
Joint effect of nitrogen sources and B vitamin supplementation of date juice [159] M.Y. Galperin, Bacterial signal transduction network in a genomic
on lactic acid production by Lactobacillus casei subsp. rhamnosus, Bioresour. perspective, Environ. Microbiol. 6 (2004) 552–567, http://dx.doi.org/10.
Technol. 96 (2005) 63–67, http://dx.doi.org/10.1016/j.biortech.2003.09.018. 1111/j.1462-2920.2004.00633.x.
[137] E. Selmer-Olsen, T. Sorhaug, Comparative studies of the growth of [160] R. Wu, W. Zhang, T. Sun, J. Wu, X. Yue, H. Meng, H. Zhang, Proteomic analysis
Lactobacillus plantarum in whey supplemented with autolysate from of responses of a new probiotic bacterium Lactobacillus casei Zhang to low
brewery yeast biomass or commercial yeast extract, Milchwissenschaft 53 acid stress, Int. J. Food Microbiol. 147 (2011) 181–187, http://dx.doi.org/10.
(1998) 367–370. 1016/j.ijfoodmicro.2011.04.003.
[138] M. Altaf, B.J. Naveena, M. Venkateshwar, E.V. Kumar, G. Reddy, Single step [161] F. Lorquet, P. Goffin, L. Muscariello, J.-B. Baudry, V. Ladero, M. Sacco, M.
fermentation of starch to L(+) lactic acid by Lactobacillus amylophilus GV6 in Kleerebezem, P. Hols, Characterization and functional analysis of the poxB
SSF using inexpensive nitrogen sources to replace peptone and yeast gene, which encodes pyruvate oxidase in Lactobacillus plantarum, J.
extract—optimization by RSM, Process Biochem. 41 (2006) 465–472, http:// Bacteriol. 186 (2004) 3749–3759, http://dx.doi.org/10.1128/JB.186.12.3749-
dx.doi.org/10.1016/j.procbio.2005.07.011. 3759.2004.
[139] B. Liu, M. Yang, B. Qi, X. Chen, Z. Su, Y. Wan, Optimizing L-(+)-lactic acid [162] P. Goffin, L. Muscariello, F. Lorquet, A. Stukkens, D. Prozzi, M. Sacco, M.
production by thermophile Lactobacillus plantarum As.1.3 using alternative Kleerebezem, P. Hols, Involvement of pyruvate oxidase activity and acetate
nitrogen sources with response surface method, Biochem. Eng. J. 52 (2010) production in the survival of Lactobacillus plantarum during the stationary
212–219, http://dx.doi.org/10.1016/j.bej.2010.08.013. phase of aerobic growth, Appl. Environ. Microbiol. 72 (2006) 7933–7940,
[140] J. Marták, Š. Schlosser, E. Sabolová, L. Krištofıı́ková, M. Rosenberg, http://dx.doi.org/10.1128/AEM.00659-06.
Fermentation of lactic acid with Rhizopus arrhizus in a stirred tank reactor [163] M.G. Murphy, L. O’Connor, D. Walsh, S. Condon, Oxygen dependet lactate
with a periodical bleed and feed operation, Process Biochem. 38 (2003) utilization by Lactobacillus plantarum, Arch. Microbiol. 141 (1985) 75–79.
1573–1583, http://dx.doi.org/10.1016/S0032-9592(03)00059-1. [164] P.A. Bron, M. Wels, R.S. Bongers, H. van Bokhorst-van de Veen, A. Wiersma,
[141] T. Pauli, J.J. Fitzpatrick, Malt combing nuts as a nutrient supplement to whey L. Overmars, M.L. Marco, M. Kleerebezem, Transcriptomes reveal genetic
permeate for producing lactic by fermentation with Lactobacillus casei, signatures underlying physiological variations imposed by different
Process Biochem. 38 (2002) 1–6, http://dx.doi.org/10.1016/S0032- fermentation conditions in Lactobacillus plantarum, PLoS One 7 (2012)
9592(02)00038-9. e38720, http://dx.doi.org/10.1371/journal.pone.0038720.
[142] L. Yu, T. Lei, X. Ren, X. Pei, Y. Feng, Response surface optimization of [165] A. Guidone, R.G. Ianniello, A. Ricciardi, T. Zotta, E. Parente, Aerobic
L-(+)-lactic acid production using corn steep liquor as an alternative metabolism and oxidative stress tolerance in the Lactobacillus plantarum
nitrogen source by Lactobacillus rhamnosus CGMCC 1466, Biochem. Eng. J. 39 group, World J. Microbiol. Biotechnol. 29 (2013) 1713–1722, http://dx.doi.
(2008) 496–502, http://dx.doi.org/10.1016/j.bej.2007.11.008. org/10.1007/s11274-013-1334-0.
[143] B. Kamm (Ed.), Microorganisms in Biorefineries, Springer, Berlin, Heidelberg, [166] S.-Q. Liu, R. Holland, V.L. Crow, Serine metabolism by a Lactobacillus
2015, http://dx.doi.org/10.1007/978-3-662-45209-7. plantarum adjunct, Aust. J. Dairy Technol. 57 (2002) 177.
[144] Lactic Acid Bacteria, in: W.H. Holzapfel, B.J.B. Wood (Eds.), John Wiley & [167] B.P. Upadhyaya, L.C. DeVeaux, L.P. Christopher, Metabolic engineering as a
Sons, Ltd, Chichester, UK, 2014, http://dx.doi.org/10.1002/9781118655252. tool for enhanced lactic acid production, Trends Biotechnol. 32 (2014)
[145] D. Agyei, M.K. Danquah, Carbohydrate utilization affects Lactobacillus 637–644, http://dx.doi.org/10.1016/j.tibtech.2014.10.005.
delbrueckii subsp. lactis 313 cell-enveloped-associated proteinase [168] E. Stefanovic, G. Fitzgerald, O. McAuliffe, Advances in the genomics and
production, Biotechnol. Bioprocess Eng. 17 (2012) 787–794, http://dx.doi. metabolomics of dairy lactobacilli: a review, Food Microbiol. 61 (2017)
org/10.1007/s12257-012-0106-2. 33–49, http://dx.doi.org/10.1016/j.fm.2016.08.009.
[146] S.-Q. Liu, R. Holland, V.L. Crow, The potential of dairy lactic acid bacteria to [169] R. Mazzoli, F. Bosco, I. Mizrahi, E.A. Bayer, E. Pessione, Towards lactic acid
metabolise amino acids via non-transaminating reactions and endogenous bacteria-based biorefineries, Biotechnol. Adv. 32 (2014) 1216–1236, http://
transamination, Int. J. Food Microbiol. 86 (2003) 257–269, http://dx.doi.org/ dx.doi.org/10.1016/j.biotechadv.2014.07.005.
10.1016/S0168-1605(03)00040-0. [170] C. Hidalgo-Cantabrana, S. O’Flaherty, R. Barrangou, CRISPR-based
[147] T. Iino, A. Manome, S. Okada, T. Uchimura, K. Komagata, Effects of sodium engineering of next-generation lactic acid bacteria, Curr. Opin. Microbiol. 37
acetate on the production of stereoisomers of lactic acid by Lactobacillus (2017) 79–87, http://dx.doi.org/10.1016/j.mib.2017.05.015.
238 R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239

[171] E. Gyo Lee, S.H. Moon, Y. Keun Chang, I.K. Yoo, H. Nam Chang, Lactic acid [194] Y. Wang, M. Wang, D. Cai, B. Wang, Z. Wang, P. Qin, T. Tan, Efficient L-lactic
recovery using two-stage electrodialysis and its modelling, J. Memb. Sci. 145 acid production from sweet sorghum bagasse by open simultaneous
(1998) 53–66, http://dx.doi.org/10.1016/S0376-7388(98)00065-9. saccharification and fermentation, RSC Adv. 6 (2016) 35771–35777, http://
[172] K.L. Wasewar, A.B.M. Heesink, G.F. Versteeg, V.G. Pangarkar, Reactive dx.doi.org/10.1039/C6RA04538C.
extraction of lactic acid using alamine 336 in MIBK: equilibria and kinetics, J. [195] S. Hama, S. Mizuno, M. Kihara, T. Tanaka, C. Ogino, H. Noda, A. Kondo,
Biotechnol. 97 (2002) 59–68, http://dx.doi.org/10.1016/S0168- Production of D-lactic acid from hardwood pulp by mechanical milling
1656(02)00057-3. followed by simultaneous saccharification and fermentation using
[173] K.L. Wasewar, V.G. Pangarkar, A.B.M. Heesink, G.F. Versteeg, Intensification metabolically engineered Lactobacillus plantarum, Bioresour. Technol. 187
of enzymatic conversion of glucose to lactic acid by reactive extraction, (2015) 167–172, http://dx.doi.org/10.1016/j.biortech.2015.03.106.
Chem. Eng. Sci. 58 (2003) 3385–3393, http://dx.doi.org/10.1016/S0009- [196] J. Hu, Y. Lin, Z. Zhang, T. Xiang, Y. Mei, S. Zhao, Y. Liang, N. Peng, High-titer
2509(03)00221-5. lactic acid production by Lactobacillus pentosus FL0421 from corn stover
[174] D. Yankov, J. Molinier, J. Albet, G. Malmary, G. Kyuchoukov, Lactic acid using fed-batch simultaneous saccharification and fermentation, Bioresour.
extraction from aqueous solutions with tri-n-octylamine dissolved in Technol. 214 (2016) 74–80, http://dx.doi.org/10.1016/j.biortech.2016.04.
decanol and dodecane, Biochem. Eng. J. 21 (2004) 63–71, http://dx.doi.org/ 034.
10.1016/j.bej.2004.03.006. [197] L. Ye, M.S. Bin Hudari, Z. Li, J.C. Wu, Simultaneous detoxification,
[175] A. Komesu, M.R. Wolf Maciel, J.A. Rocha de Oliveira, L.H. da Silva Martins, R. saccharification and co-fermentation of oil palm empty fruit bunch
Maciel Filho, Purification of lactic acid produced by fermentation: focus on hydrolysate for L-lactic acid production by Bacillus coagulans JI12, Biochem.
non-traditional distillation processes, Sep. Purif. Rev. 46 (2017) 241–254, Eng. J. 83 (2014) 16–21, http://dx.doi.org/10.1016/j.bej.2013.12.005.
http://dx.doi.org/10.1080/15422119.2016.1260034. [198] F. Demichelis, D. Pleissner, S. Fiore, S. Mariano, I.M. Navarro Gutiérrez, R.
[176] Y.H. Kim, S.-H. Moon, Lactic acid recovery from fermentation broth using Schneider, J. Venus, Investigation of food waste valorization through
one-stage electrodialysis, J. Chem. Technol. Biotechnol. 76 (2001) 169–178, sequential lactic acid fermentative production and anaerobic digestion of
http://dx.doi.org/10.1002/jctb.368. fermentation residues, Bioresour. Technol. 241 (2017) 508–516, http://dx.
[177] A. Komesu, P.F.M. Martinez, B.H. Lunelli, R.M. Filho, M.R.W. Maciel, Lactic doi.org/10.1016/j.biortech.2017.05.174.
acid purification by reactive distillation system using design of experiments, [199] D. Pleissner, F. Demichelis, S. Mariano, S. Fiore, I.M. Navarro Gutiérrez, R.
Chem. Eng. Process. Process Intensif. 95 (2015) 26–30, http://dx.doi.org/10. Schneider, J. Venus, Direct production of lactic acid based on simultaneous
1016/j.cep.2015.05.005. saccharification and fermentation of mixed restaurant food waste, J. Clean.
[178] J. Marták, Š. Schlosser, Liquid-liquid equilibria of butyric acid for solvents Prod. 143 (2017) 615–623, http://dx.doi.org/10.1016/j.jclepro.2016.12.065.
containing a phosphonium ionic liquid, Chem. Pap. 62 (2008), http://dx.doi. [200] L. Zhang, X. Li, Q. Yong, S.-T. Yang, J. Ouyang, S. Yu, Simultaneous
org/10.2478/s11696-007-0077-5. saccharification and fermentation of xylo-oligosaccharides manufacturing
[179] H.-J. Huang, S. Ramaswamy, U.W. Tschirner, B.V. Ramarao, A review of waste residue for L-lactic acid production by Rhizopus oryzae, Biochem. Eng.
separation technologies in current and future biorrfineries, Sep. Purif. J. 94 (2015) 92–99, http://dx.doi.org/10.1016/j.bej.2014.11.020.
Technol. 62 (2008) 1–21, http://dx.doi.org/10.1016/j.seppur.2007.12.011. [201] I.-Y. Eom, Y.-H. Oh, S.J. Park, S.-H. Lee, J.-H. Yu, Fermentative L-lactic acid
[180] J. Sikder, S. Chakraborty, P. Pal, E. Drioli, C. Bhattacharjee, Purification of production from pretreated whole slurry of oil palm trunk treated by
lactic acid from microfiltrate fermentation broth by cross-flow hydrothermolysis and subsequent enzymatic hydrolysis, Bioresour. Technol.
nanofiltration, Biochem. Eng. J. 69 (2012) 130–137, http://dx.doi.org/10. 185 (2015) 143–149, http://dx.doi.org/10.1016/j.biortech.2015.02.060.
1016/j.bej.2012.09.003. [202] M. Smerilli, M. Neureiter, S. Wurz, C. Haas, S. Frühauf, W. Fuchs, Direct
[181] X. Sun, Q. Wang, W. Zhao, H. Ma, K. Sakata, Extraction and purification of fermentation of potato starch and potato residues to lactic acid by
lactic acid from fermentation broth by esterification and hydrolysis method, Geobacillus stearothermophilus under non-sterile conditions, J. Chem.
Sep. Purif. Technol. 49 (2006) 43–48, http://dx.doi.org/10.1016/j.seppur. Technol. Biotechnol. Publ. 90 (2015) 648–657, http://dx.doi.org/10.1002/
2005.08.005. jctb.4627.
[182] J.M.K. Timmer, J. Kromkamp, T. Robbertsen, Lactic acid separation from [203] S. Cingadi, K. Srikanth, E.V.R. Arun, S. Sivaprakasam, Statistical optimization
fermentation broths by reverse osmosis and nanofiltration, J. Memb. Sci. 92 of cassava fibrous waste hydrolysis by response surface methodology and
(1994) 185–197, http://dx.doi.org/10.1016/0376-7388(94)00061-1. use of hydrolysate based media for the production of optically pure D-lactic
[183] J.M.K. Timmer, H.C. van der Horst, T. Robbertsen, Transport of lactic acid acid, Biochem. Eng. J. 102 (2015) 82–90, http://dx.doi.org/10.1016/j.bej.
through reverse osmosis and nanofiltration membranes, J. Memb. Sci. 85 2015.02.006.
(1993) 205–216, http://dx.doi.org/10.1016/0376-7388(93)85169-W. [204] J. Won, J. Hoon, J. Park, J. Shin, J. Hwan, B. Hyun, J. Sohn, J. Seo, J. Park, S. Rin,
[184] X. Wang, Y. Wang, X. Zhang, H. Feng, T. Xu, In-situ combination of D. Kweon, Co-expression of two heterologous lactate dehydrogenases genes
fermentation and electrodialysis with bipolar membranes for the in Kluyveromyces marxianus for L-lactic acid production, J. Biotechnol. 241
production of lactic acid: continuous operation, Bioresour. Technol. 147 (2017) 81–86, http://dx.doi.org/10.1016/j.jbiotec.2016.11.015.
(2013) 442–448, http://dx.doi.org/10.1016/j.biortech.2013.08.045. [205] T.L. Turner, E. Kim, C. Hwang, G. Zhang, J. Liu, Y.-S. Jin, Short communication:
[185] L. Wang, B. Zhao, F. Li, K. Xu, C. Ma, F. Tao, Q. Li, P. Xu, Highly efficient conversion of lactose and whey into lactic acid by engineered yeast, J. Dairy
production of D-lactate by Sporolactobacillus sp. CASD with simultaneous Sci. 100 (2017) 124–128, http://dx.doi.org/10.3168/jds.2016-11784.
enzymatic hydrolysis of peanut meal, Appl. Microbiol. Biotechnol. 89 (2011) [206] S. Baek, E.Y. Kwon, S. Bae, B. Cho, S. Kim, J. Hahn, Improvement of D-lactic
1009–1017, http://dx.doi.org/10.1007/s00253-010-2904-9. acid production in Saccharomyces cerevisiae under acidic conditions by
[186] Y. Meng, Y. Xue, B. Yu, C. Gao, Y. Ma, Efficient production of L-lactic acid with evolutionary and rational metabolic engineering, Biotechnol. J. 12 (2017)
high optical purity by alkaliphilic Bacillus sp. WL-S20, Bioresour. Technol. 1700015, http://dx.doi.org/10.1002/biot.201700015.
116 (2012) 334–339, http://dx.doi.org/10.1016/j.biortech.2012.03.103. [207] S. Baek, E.Y. Kwon, Y.H. Kim, J. Hahn, Metabolic engineering and adaptive
[187] Y.-C. Kuo, S.-F. Yuan, C.-A. Wang, Y.-J. Huang, G.-L. Guo, W.-S. Hwang, evolution for efficient production of D-lactic acid in Saccharomyces
Production of optically pure L-lactic acid from lignocellulosic hydrolysate by cerevisiae, Appl. Microbiol. Biotechnol. 100 (2016) 2737–2748, http://dx.doi.
using a newly isolated and D-lactate dehydrogenase gene-deficient org/10.1007/s00253-015-7174-0.
Lactobacillus paracasei strain, Bioresour. Technol. 198 (2015) 651–657, [208] R. Yamada, K. Wakita, R. Mitsui, H. Ogino, Enhanced D-lactic acid production
http://dx.doi.org/10.1016/j.biortech.2015.09.071. by recombinant Saccharomyces cerevisiae following optimization of the
[188] Y. Wang, Z. Yang, P. Qin, T. Tan, Fermentative L-(+)-lactic acid production global metabolic pathway, Biotechnol. Bioeng. 114 (2017) 2075–2084,
from defatted rice bran, RSC Adv. 4 (2014) 8907, http://dx.doi.org/10.1039/ http://dx.doi.org/10.1002/bit.26330.
c3ra46140h. [209] R.A. Weusthuis, A.E. Mars, J. Springer, E.J. Wolbert, H. van der Wal, T.G. de
[189] Z. Shi, P. Wei, X. Zhu, J. Cai, L. Huang, Z. Xu, Efficient production of L-lactic Vrije, M. Levisson, A. Leprince, G.B. Houweling-Tan, A.P.H.A. Moers, S.N.
acid from hydrolysate of Jerusalem artichoke with immobilized cells of Hendriks, O. Mendes, Y. Griekspoor, M.W. Werten, P.J. Schaap, J. van der
Lactococcus lactis in fibrous bed bioreactors, Enzyme Microb. Technol. 51 Oost, G. Eggink, Monascus ruber as cell factory for lactic acid production at
(2012) 263–268, http://dx.doi.org/10.1016/j.enzmictec.2012.07.007. low pH, Metab. Eng. 42 (2017) 66–73, http://dx.doi.org/10.1016/j.ymben.
[190] X.-Y. Ge, Enhancement of L-lactic acid production in Lactobacillus casei from 2017.05.005.
Jerusalem artichoke tubers by kinetic optimization and citrate metabolism, [210] Y. Wang, K. Li, F. Huang, J. Wang, J. Zhao, X. Zhao, E. Garza, R. Manow, S.
J. Microbiol. Biotechnol. 20 (2010) 101–109, http://dx.doi.org/10.4014/jmb. Grayburn, S. Zhou, Engineering and adaptive evolution of Escherichia coli W
0905.05032. for L-lactic acid fermentation from molasses and corn steep liquor without
[191] S. Gandolfi, L. Pistone, G. Ottolina, P. Xu, S. Riva, Hemp hurds biorefining: a additional nutrients, Bioresour. Technol. 148 (2013) 394–400, http://dx.doi.
path to green L-(+)-lactic acid production, Bioresour. Technol. 191 (2015) org/10.1016/j.biortech.2013.08.114.
59–65, http://dx.doi.org/10.1016/j.biortech.2015.04.118. [211] S. Angermayr, A.D. van der Woude, D. Correddu, A. Vreugdenhil, V. Verrone,
[192] Z. Zhang, Y. Xie, X. He, X. Li, J. Hu, Z. Ruan, S. Zhao, N. Peng, Y. Liang, K.J. Hellingwerf, Exploring metabolic engineering design principles for the
Comparison of high-titer lactic acid fermentation from NaOH- and photosynthetic production of lactic acid by Synechocystis sp. PCC6803,
NH3 -H2 O2 -pretreated corncob by Bacillus coagulans using simultaneous Biotechnol. Biofuels 7 (2014) 99, http://dx.doi.org/10.1186/1754-6834-7-99.
saccharification and fermentation, Sci. Rep. 6 (2016) 37245, http://dx.doi. [212] K. Okano, S. Yoshida, T. Tanaka, C. Ogino, H. Fukuda, A. Kondo,
org/10.1038/srep37245. Homo-D-lactic acid fermentation from arabinose by redirection of the
[193] S.G. Karp, A.H. Igashiyama, P.F. Siqueira, J.C. Carvalho, L.P.S. Vandenberghe, phosphoketolase pathway to the pentose phosphate pathway in L-lactate
V. Thomaz-Soccol, J. Coral, J.-L. Tholozan, A. Pandey, C.R. Soccol, Application dehydrogenase gene-deficient Lactobacillus plantarum, Appl. Environ.
of the biorefinery concept to produce L-lactic acid from the soybean vinasse Microbiol. 75 (2009) 5175–5178, http://dx.doi.org/10.1128/AEM.00573-09.
at laboratory and pilot scale, Bioresour. Technol. 102 (2011) 1765–1772, [213] K. Okano, Q. Zhang, S. Shinkawa, S. Yoshida, T. Tanaka, H. Fukuda, A. Kondo,
http://dx.doi.org/10.1016/j.biortech.2010.08.102. Efficient production of optically pure D-lactic acid from raw corn starch by
R. Alves de Oliveira et al. / Biochemical Engineering Journal 133 (2018) 219–239 239

using a genetically modified L-lactate dehydrogenase gene-deficient and Global lactic acid market size, share, growth, outlook and
-amylase-secreting Lactobacillus plantarum strain, Appl. Environ. Microbiol. forecast to 2022: Acute Market Reports. Available at: http://www.
75 (2009) 462–467, http://dx.doi.org/10.1128/AEM.01514-08.
[214] Y. Zhang, A. Kumar, P.R. Hardwidge, T. Tanaka, A. Kondo, P.V. Vadlani, wtva.com/story/35918467/global-lactic-acid-market-size-share-
D-lactic acid production from renewable lignocellulosic biomass via growth-outlook-and-forecast-to-2022-acute-market-reports. July
genetically modified Lactobacillus plantarum, Biotechnol. Prog. 32 (2016) 19, 2017. Accessed: August 1st, 2017.
271–278, http://dx.doi.org/10.1002/btpr.2212.
[215] A. Komesu, P.F. Martins, B.H. Lunelli, J.O. Rocha, R. Maciel Filho, M.R. Wolf
New market research projects strong growth for lactic
Maciel, The effect of evaporator temperature on lactic acid purity and acid. Available at: http://www.bioplasticsmagazine.com/en/news/
recovery by short path evaporation, Sep. Sci. Technol. 50 (2015) 1548–1553, meldungen/new-market-study-lactic-acid.php. August 8, 2014.
http://dx.doi.org/10.1080/01496395.2014.975363.
Accessed: August 1st, 2017.
[216] A. Komesu, P.F. Martins, B.H. Lunelli, J. Oliveira, R. Maciel Filho, M.R. Wolf
Maciel, Evaluation of lactic acid purification from fermentation broth by Products releases. Available at: https://www.lactic.com/en-us/
hybrid short path evaporation using factorial experimental design, Sep. news/productreleases.aspx#195-2017. Accessed: August 1st, 2017.
Purif. Technol. 136 (2014) 233–240, http://dx.doi.org/10.1016/j.seppur.
Global polylactic acid (PLA) market 2015–2019 with BASF,
2014.09.010.
[217] D. Pleissner, R. Schneider, J. Venus, T. Koch, Separation of lactic acid and Braschem, Eastman Chemicals, NatureWorks, Nova Chemicals &
recovery of salt-ions from fermentation broth, J. Chem. Technol. Biotechnol. Sinopec Dominating. Available at: http://www.prnewswire.com/
92 (2017) 504–511, http://dx.doi.org/10.1002/jctb.5023. news-releases/global-polylactic-acid-pla-market-2015-2019-
[218] C. Wang, Q. Li, D. Wang, J. Xing, Improving the lactic acid production of
Actinobacillus succinogenes by using a novel fermentation and separation with-basf-braschem-eastman-chemicals-natureworks-nova-
integration system, Process Biochem. 49 (2014) 1245–1250, http://dx.doi. chemicals-sinopec-dominating-300055251.html. March 24, 2015.
org/10.1016/j.procbio.2014.04.009. Accessed: August 1st, 2017.
[219] P. Pal, P. Dey, Process intensification in lactic acid production by three stage
membrane integrated hybrid reactor system, Chem. Eng. Process. Process Lactic acid: the lifeblood of our business. Available at: http://
Intensif. 64 (2013) 1–9, http://dx.doi.org/10.1016/j.cep.2012.12.006. www.corbion.com/about-corbion/corbion-stories/lactic-acid-the-
[220] H.D. Lee, M.Y. Lee, Y.S. Hwang, Y.H. Cho, H.W. Kim, H.B. Park, Separation and lifeblood-of-our-business. Accessed: August 1st, 2017 (A).
purification of lactic acid from fermentation broth using
membrane-integrated separation processes, Ind. Eng. Chem. Res. 56 (2017)
Factbox. Available at: http://www.corbion.com/about-corbion/
8301–8310, http://dx.doi.org/10.1021/acs.iecr.7b02011. corbion-stories/lactic-acid-the-lifeblood-of-our-business/under-
[221] P. Khunnonkwao, P. Boontawan, D. Haltrich, T. Maischberger, A. Boontawan, the-microscope-lactic-acid. Accessed: August 1st, 2017 (B).
Purification of L-(+)-lactic acid from pre-treated fermentation broth using
The appliance of science. Available at: http://www.corbion.com/
vapor permeation-assisted esterification, Process Biochem. 47 (2012)
1948–1956, http://dx.doi.org/10.1016/j.procbio.2012.07.011. about-corbion/innovation. Accessed: August 1st, 2017 (C).
[222] J. Yu, A. Zeng, X. Yuan, X. Zhang, J. Ju, Optimizing and scale-up strategy of CORBION. Available at: https://www.euronext.com/en/
molecular distillation for the purification of lactic acid from fermentation
products/equities/NL0010583399-XAMS/company-information.
broth, Sep. Sci. Technol. 6395 (2015), http://dx.doi.org/10.1080/01496395.
2015.1056363, 150623131312002. Accessed: August 1st, 2017.
About us. Available at: https://www.lactic.com/en-us/aboutus.
aspx. Accessed: August 1st, 2017 (A).
WEB references Products releases. Available at: https://www.lactic.com/en-us/
news/productreleases.aspx#195-2017. Accessed: August 1st, 2017
Lactic acid – Global strategic business report 2017 – Poised for
(B).
phenomenal growth. Available at: http://markets.businessinsider.
About NatureWorks. Available at: http://www.natureworksllc.
com/news/stocks/Lactic-Acid-Global-Strategic-Business-Report-
com/About-NatureWorks. Accessed: August 1st, 2017.
2017-Poised-for-Phenomenal-Growth-511670. July 26, 2017.
NatureWorks LLC company details. Available at: https://www.
Accessed: August 1st, 2017.
industryintel.com/public:i2topic/company/NatureWorks%20LLC.
Pharma Compass: Lactic Acid > Indian Import/Export Trade Data
Accessed: August 1st, 2017.
> API Reference Price. Available at: https://www.pharmacompass.
2017-2022 global lactic acid & polylactic acid market analysis:
com/price/lactic-acid. Accessed: December, 19 th, 2017.
Galactic, B&G, ADM. Available at: https://firstnewshawk.com/
Lactic acid market size worth $9.8bn by 2025 & PLA to reach
2017-2022-global-lactic-acid-polylactic-acid-market-analysis/.
$6.5bn. Available at: http://www.grandviewresearch.com/press-
July 24, 2017. Accessed: August 1st, 2017.
release/global-lactic-acid-and-poly-lactic-acid-market. May 2017.
ISO 14044:2006. Environmental management - Life cycle assess-
Accessed: August 1st, 2017.
ment - Requirements and guidelines. Available at: https://www.iso.
org/standard/38498.html. Accessed: November 16 th, 2017.

You might also like