throbber
PETITIONERS
`
`EXHIBIT NO. 1019 Page 1 of 35
`
`

`

`PETITIONERS
`
`EXHIBIT NO. 1019 Page 2 of 35
`
`

`

`148
`
`Biosurfactants: Research Trends and Applications
`
`Surfactants are classified as cationic, anionic, zwitterionic, and nonionic and are
`
`Some surfactants, known as biosurfactants, are biologically produced from yeast
`or bacteria (Lin, 1996). They can be potentially as effective with some distinct
`advantages over the highly used synthetic surfactants due to high specificity, biode-
`gradability, and biocompatibility (Cooper, 1986).
`Biosurfactants are grouped as glycolipids, lipopeptides, phospholipids, fatty acids,
`and neutral lipids (Bierman et al., 1987). Most of these compounds are either anionic
`or neutral, with only a few cationic ones. The hydrophobic parts of the molecule are
`based on long-chain fatty acids, hydroxy fatty acids, or or-alkyl—p-hydroxy fatty acids.
`The hydrophilic portion can be a carbohydrate, amino acid, cyclic peptide, phosphate,
`carboxylic acid, or alcohol. A wide variety of microorganisms can produce these
`compounds. The CMCs of the biosurfactants generally range from 1 to 200 mg/L
`and their moiecular weights (MWS) from 500 to 1500 amu (Lang and Wagner, 1987).
`
`made synthetically from hydrocarbons, lignosulfonates, or triglycerides. Some com—
`mon synthetic surfactants include linear alkyl benzenesulfonates, alcohol sulfates,
`alcohol ether sulfates, alcohol glyceryl ether suifonates, oc~olefin sulfonates, alcohol
`ethoxylates, and alkylphenol ethoxylates (Layman, 1985). Surfactants have many
`applications industrially with multiphasic systems. Sodium dodecyl sulfate (SDS,
`CI2H25~SO4' Nat) is a widely used anionic surfactant. The effectiveness of a surfac-
`tant is determined by surface tension lowering, which is a measure of the surface free
`energy per unit area or the work required to bring a molecule from the bulk phase to
`the surface (Rosen, 1978). These amphiphilic compounds (containing hydrophobic
`and hydrophilic portions) concentrate at solid—liquid, liquid—liquid, or vapor—liquid
`interfaces. An interfacial boundary exists between two immiscible phases. The
`hydrophobic portion concentrates at the surface while the hydrophilic is oriented
`toward the solution. A good surfactant can lower the surface tension of water from 72
`to 35 mN/m and the interfacial tension (tension between nonpolar and polar liquids)
`for water against n-hexadecane from 40 to l mN/m. Efficient surfactants have a
`low CMC (i.e., less surfactant is necessary to decrease the surface tension} as the
`CMC is defined as the minimum concentration necessary to initiate micelle forma—
`tion (Becher, 1965). In practice, the CMC is also the maximum concentration of sur-
`factant monomers in water and is influenced by pH, temperature, and ionic strength.
`An important factor in the choice of surfactant is the product cost (Mulligan and
`Gibbs, 1993). In general, surfactants are used to save energy and consequently energy
`costs (such as the energy required for pumping or mixing). Charge type, physico—
`chemical behavior, solubility, and adsorption behavior are some important selection
`criteria for surfactants.
`
`etal., 2006), D (Zhao et al., 2010), bacillomycin (Roongsawang et al., 2003), fengycin
`
`LIPOPEPTIDE BIOSURFACTANTS
`
`Lipopeptides are produced by a variety of microorganisms, including Bacillus,
`Lacrobacilins, Srrepromyces, Pseudomonas, and Serratia (Cameotra and Makkar,
`
`2004; Georgiou et al., 1992). The lipopeptides are cyclic peptides with a fatty acyl
`chain. Various lipopeptides include surfactin (Roongsawang et al., 2003; Youssef
`et al., 2007),1ichenysin A (Yakimov et al., 1995) or C (Jenny et al., 1991), B (Folmsbee
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 3 of 35
`
`

`

`Characterization, Production, and Applications of Lipopeptides
`
`149
`
`Surfactin A
`
`CH3“ CH (CH2)? — CH — CH2 — CO * Glu — Leu — Leo \
`CH3
`
`Leu — Leu Asp
`
`Surfactin B
`
`CHai CH (CH2)S — CH7 CH2 — CO 7 Glu - Leu — Leu
`
`CH3
`
`SurfactinC
`
`Leu e Leo fl Asp
`
`(Vanittanakom and Loeffier, 1986), and iturin (Bonrnatin et al., 2003). Surfactin is a
`cyclic heptapeptide, with antibacterial, antifungal, antiviral, and antitumor activities
`(Folrnsbee et al., 2006; Zhao et a1... 2010).
`Lipopeptides have been tested in enhanced oil recovery and the transportation of
`crude oils (Hayes et al., 1986). They were demonstrated to be effective for antimi-
`crobial activity and in the reduction of the interfacial tension of oil and water and the
`viscosity of the oil, the removal of water from the emulsions prior to processing, and
`the release of bitumen from oil sands. Although most biosurfactant—producing organ-
`isms are aerobic, a few anaerobic producers exist. Bacillus iichemformis JFu2 is an
`example, which would be well suited for in situ studies for enhanced oil recovery or
`soil decontamination (Javaheri et al., 1985).
`Surfactin is the most studied lipopeptide and consists of a seven-amino acid
`sequence in a cyclical structure with a 13wl6 carbon fatty acid (Kakinurna et al., 1969)
`and has two charged amino acids (glutamic and aspartic acids). In addition to sur-
`factin, iturins and fengycins are also produced (Deleu et al., 1999). Their structures
`are shown in Figures 6.1 through 6.3. Iturins are cyclic peptides with seven amino
`acids and a B«amino closure. Fengycin lipopeptides are B-hydroxy fatty acids with
`an eight-member ring in an N-terminal decapeptide. At the therrninal end, there is a
`tyrosine residue at position 3. This forms an eight-member lactone ring. Fengycin A
`and B vary at position 6. The A form has an Ala compared to the B form of a valine.
`
`Bioresomr. Techno!” 101, 6118, ZOlU.)
`
`CH3— CH (CH2er —' CH "- CH2 W CO — Glu — Leu — Leu
`
`lU
`
`wH—mu e Leu — Asp
`
`Surfactin D
`
`CH3— CH (Cl-12)1U e CH- CHZ— C0 * Giu - Leo — Leu
`
`CH3
`
`OW"— Leu — Lea ~ Asp
`
`FIGURE 6.1
`
`Structures of various forms of surfactin. (Adapted from Janeir, T. et £11.,
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 4 of 35
`
`

`

`Peng et al. (2008)
`
`The fatty acid normally varies from 14 to 18 carbons in length (Arirna et al., 1968;
`Matsuyama et al., 1992; Roongsawang et al., 2003). Fengycin has three charged
`amino acids (two glutamic acids and an ornithine). The CMC is 6.25 mglL.
`Other lipopeptides have also been studied. They include lichenysin from
`B. lichem'formis (Grangemard et al., 1999; Horowitz et al., 1990), arthrofactin from
`Arthmbacter sp. (now known as Pseudomonas sp. M1838) (Morikawa et al., 1993;
`Roongsawang et al., 2003), pumilacidin from P. pumilus (Naruse et al., 1990), and
`serrawettin from Serratia marcescens (Matsuyarna et al., 1992). Others include mas-
`setolide A (Sen and Swaminathan, 1997), putisolvins I and II (Kuiper et al., 2004).
`and pumilacidin (Naruse et al., 1990). As can be seen, there is no generally accepted
`nomenclature based on the structure. Pseudomonas lipopeptides include viscosin,
`amphisin, tolassin, and syringomycin (Raaijmakers et al., 2006). Viscosin has nine
`amino acids with a 3-hydroxy fatty acid, whereas amphisin has 11 amino acids
`linked to a similar fatty acid (Soresen et al., 2001). A comparison of the CMC of
`some lipopeptides is shown in Table 6.1.
`
`— CH (CH2)12_1S — co — Glu — Orn
`
`CH3
`
`Ile — O—rw Tyr — AlloThr
`
`/
`Tyr
`\ Gln —— Pro w Ala — Glu
`
`FIGURE 6.3 Structure fengycin.
`
`W T
`
`ABLE 6.1
`
`CMC Values of Various Isolated Lipopeptides
`
`Lipopeptide
`Fengycin
`
`Microbial Source
`B. circulans DMS-2
`
`CMC (mg/L)
`10—13
`
`Surfactin
`Surfactin and fengycin
`Biosurfactant
`Lipopeptide
`
`B. subtilis
`B. subriiis
`B. Iichenifonnis
`Rhodocaccus 5p.
`
`17
`l0 and 11
`0.6
`23.?
`
`Reference
`Sivapathasekaran et al.
`(2009, 2050)
`
`Deleu et al. {1999)
`Lin et a1. (1998)
`Barres et al. (2008)
`
`Biosurfactants: Research Trends and Applications
`
`Iturin
`
`CH3- CH (CH2}9_14 r CH- CH2 — CO -’ ASn — Tyr 7 Asn \
`1
`/
`
`Set — Asn ‘- Pro
`
`FIGURE 6.2
`
`Structure of iturin.
`
`Fengycin
`
`H o
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 5 of 35
`
`

`

`tiitaracterization, Production, and Applications of Lipopeptides
`
`- 151
`
`Synthesis of the lipopeptides is performed based on a series of enzymes for each
`W'or amino acid additioa, ring closure, and acylation (Peypoux et al., 1999). This
`makes genetic manipulation difficult for enhanced production. Most studies are con-
`;cntrated on grewth optimization and isolation of overproducers (Peypoux et al.,
`1099') With the exception of a few studies (Gu et al., 2007', Nakayama et al., 1997;
`Uhno et al., 1995; Peypoux et al., 1999).
`Most of the fecus has been on higher-priced applications due to the low yields and high
`gust of the media. Higher-volume low applications including environmental remediation,
`enhanced oil rec0very, laundry soaps, and polymerization of emulsions, need the develop-
`ment of low-cost substrates such as agroindustrial wastes (Makkar and Cameotra, 1999;
`N-Iukherjee et al., 2006) and isolation techniques, bioreactor design, and higher yields.
`
`SURFACTIN
`
`B. subtilis produces surfactin, one of the first lipopeptides found in 1968 (Figure 6.1).
`It has seven amino acids bonded to the carboxyl and hydroxyl groups of a l4~carbon
`acid (Kakinuma et al., 1969) and has blood clotting properties. Surfactin concentra-
`tions as low as 0.005% reduce the surface tension to 27 mN/m, making it a powerful
`hiosurfactant. The CMC can be as low as 10 mg/L (Dae et al., 2006). The interfacial
`tension for hydrocarbon—water interfaces can be less than 1 rnN/rn.
`The primary structure of surfactin was determined many years ago by Kaldnurna et al.
`(1969). It is a heptapeptide with a B—hydroxy fatty acid within a lactone ring structure. The
`seven amino acids are bonded to the carboxyl and hydroxyl groups of a 14—carbon acid.
`More recently, the three-dimensional structure was determined by 1H NMR techniques
`(Bonrnatin et al., 1995). Surfactin folds into a b—sheet structure, which resembles a horse
`saddle both in aqueous solutions and at the air/water interface (Ishigami et al., 1995). The
`solubility and surface-active properties of the surfactin are dependent on the orientation
`of the residues. The fatty acyl chain with the hydrophobic residues formed one face,
`while the two carboxylic acid side chains form a claw structure enabling the chelation of
`heavy metals (Bonmatin et al., 1994; Gallet et al., 1999; Magetdana and Ptak, 1992). This
`property has been evaluated in soil remediation studies (Mulligan et al., 1999).
`Mixtures of surfactin produced by B. subtilis have been characterized by mass
`spectrometry (Hue et al., 2001). A combination of liquid—secondary ion mass spec-
`trometry (LSI-MS) and high—energy tandem mass spectrometry (MS/MS) showed
`that the amino acid composition or length of the acyl chain can vary from 12 to
`16 carbons. Leucine and isoleucine can also be differentiated. Data obtained from
`protonated and cationized fragments were also useful for structural characterization.
`They are known as A, B, C, and D forms (Figure 6.1).
`
`
`
`SURFACTIN PRODUCTION
`
`Most biosurfactants are produced from hydrocarbon substrates (Syldatk and Wagner,
`1987). Production can be growth associated. In this case, they can either use the emulsi-
`ficaticn of the substrate (extracellular) or facilitate the passage of the substrate through
`the membrane (cell membrane associated). Biosurfactants, however, are also pro-
`duced from carbohydrates, which are very soluble. Gram-positive and gram-negative
`
`m a! u;
`
`nun-cunt In)Mimi; '
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 6 of 35
`
`

`

`1152
`
`Biosurfactants: Research Trends and Applications
`
`bacteria can produce cyclic lipopeptides. The different structures can lead to different
`properties. The biosurfactants have been postulated to enhance the growth on hydro-
`carbons and, in this case, may influence the ecology of the host sponge.
`Solid-state fermentation using okara, a soybean curd residue, has been performed
`by Ohno et al. (1995). Other substrates studied have included starch (Sandrin et al.,
`1990), cassava waste (Santos et al., 2000), molasses (Makkar and Cameotra, 1997b),
`soybean (Kim et al., 2009), and potato wastes (Fox and Bala, 2000). Surfactin yields
`from an autoclaved purified starch were 0.154 g/g. Low solid potato effluents exhib-
`ited a 66% lower surfactin yield than the purified starch (Thompson et al., 2001).
`It has been postulated that higher yields result from nutritional limitations.
`, Surfactin yields during production are low (0.02 g/g glucose) (de Roubin et al., 1989).
`Addition of iron and manganese can enhance concentrations to 0.7 g/L (Rosenberg, 1986).
`Further work on iron addition performed by Wei and Chu (1998) determined that addition
`of 1.7 mM of iron can lead to the production of up to 3.5 g/L of surfactin and enhanced
`biomass production. Alkaline addition is required to overcome the decrease in pH to
`below 5 due to acid formation. Further studies by Wei and Chu (2002) showed the effect
`of manganese on nitrogen utilization and subsequently surfactin production. A 0.1 mM
`magnesium sulfate concentration increased almost ninefold the surfactin level to 2.6 g/L.
`Wei et a1. (2007) subsequently used the Taguchi method to optimize surfactin production
`with regard to the presence of Mg, K, Mn, and Fe. They found that K and Mg were criti-
`cal. Kinsinger et al. (2003, 2005) further determined that concentrations of the four ions
`
`high concentrations of NaCl (up to 4%) and pH values from 4.5 to 10.5.
`
`could be optimized and allowed the production of 3.34 g/L of surfactin.
`Yields of 0.14 g/g sugar have been obtained using peat as a substrate after hydro-
`lysis with 0.5% sulfuric acid for 1 h at 120°C (Sheppard and Mulligan, 1987). Citric
`acid addition to glucose media could also enhance production (de Roubin et al., 1989).
`In attempts to influence the metabolic pathway, glutamic acid, leucine, aspartic acid,
`and valine were added to the media but did not enhance production. Nitrogen, how-
`ever, was a significant factor in surfactin production. Doubling anunonium nitrate
`concentrations from 0.4% to 0.8% increased yields by a factor of 1.6, while organic
`nitrogen addition did not have any benefit.
`Other investigators (Davis et al., 1999) found that surfactin yields were high-
`est in nitrate~1imited oxygen-depleted conditions, followed by ammonium-limited
`(0.075 g surfactin per g biomass), oxygen-depleted conditions (0.012 g/g biomass),
`and carbon-limited, oxygen—depleted conditions (0.0069 g/g biomass).
`A strain of B. subrilis was able to produce biosurfactant at 45°C at high NaCl con—
`centrations (4%) and a wide pH range (45—105) (Makkar and Cameotra, 1997a,b).
`It was able to remove 62% of the oil in a sand pack saturated with kerosene and thus
`could be used for in situ oil removal and cleaning sludge from sludge tanks.
`Makkar and Cameotra (2002) studied another strain of B. subtilis MTCC2423.
`They found it preferred sodium or potassium nitrate (3 g/L) or urea (1 g/L).
`Magnesium concentrations of 2.43 mM and calcium concentrations of 0.36 mM
`were optimal for biosurfactant yield. Unlike for previous studies for B. sabtilis by
`de Roubin et al. (1989), aspartic acid, asparagine, glutamic acid, valine, and lysine
`increased biosurfactant production by 60%. While glycine and leucine addition had
`no affect, alanine and arginine decreased production. Production was good even at
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 7 of 35
`
`

`

`Characterization, Production, and Applications of Lipopeptides
`
`153
`
`Solid carriers have also been evaluated for surfactin yield enhancement (Yeh et a1.,
`2005). Activated carbon and expanded clay were added at concentrations of 133 g/L.
`Surfactin at concentrations of 2150 and 3300 mgi’L were obtained for each carrier,
`resPectively. Activated carbon was more appropriate for the fermentation process and
`seemed to increase cell growth and thus yield. A summary of the yields of surfactin
`can be seen in Table 6.2.
`
`Das et a1. (2009) determined that antimicrobial activity was obtained from a
`glucose substrate, instead of sucrose, starch. and glycerol. Emulsifying lipopeptide
`biosurfactants from Azotobacter chroocaccum can be produced from oil (crude,
`waste motor lubricant) and peanut oil cake (Thavasi et 211., 2009).
`Production of another lipopeptide, brevifactin, was characterized and opti—
`mized by the marine strain Brevibacterium anreum MSA13 (Kiran et al., 2010).
`
`
`
`TABLE 6.2
`
`Production of Surfactin
`
`B. subfilis Strain
`
`ATCC 21332
`
`R1314
`
`Substrate
`
`Synthetic or scmisynthetic
`peat hydrolysate
`Aqueous two phase
`
`M1113 (pc12)
`M1113 (pC12)d
`ATCC 55033
`
`Mutant strain of
`ATCC 21332
`
`C9 (KCTC 370113)
`ATCC 21332
`ATCC 21332
`
`ATCC 21332
`
`MTCC 2423
`SD 901
`ATCC 21332
`Isolate
`
`Semisynthetic
`
`Solid-state okara
`
`Semisynthetic
`Solid-state okara
`
`Semisynthetic
`
`Sy nthetic
`
`Glucose with modified salts
`and oxygen limitation
`Glucose and mineral salts
`with iron
`
`Glucose with oxygen and
`nitrogen depletion
`Purified starch
`Sucrose with mineral salts
`
`Bean extract
`Solid carriers
`Sucrose with foam collection
`
`—Lu__,—___~_—___H__—_
`
`Surfactin Yield 0r
`Concentration
`
`100—250 rug/L
`
`160 ingi'L
`350 mg/L
`250 mg/L
`
`200-250 rug/kg
`wet mass
`
`350 mg/L
`2000 mgi'kg wet mass
`
`3500—4300 mgi‘L
`2000—4000 mg/L
`550 IngfL
`7.0 gi'L
`
`3.5 gi'L
`
`0.44 g/L
`
`0.154 gig
`1.23 glL
`3000750000 mglL
`
`21503300 mg/L
`0,25 gig
`
`Reference
`
`Arirna et a1. (1968):
`Cooper et a1. (1981)
`Sheppard and
`Mulligan (1987)
`Drouin and
`
`Cooper (1992)
`Ohno et al. (1992)
`Ohno et a1. (1992)
`
`Ohno et a1. (1992)
`Ohno et a1. (1992)
`Carrera et a]. (1992)
`Camera et al. (1993a,b)
`Mulligan et a]. (1989)
`Kim et all (1997)
`
`Wei and)ChLl (1998)
`
`Davis et a]. (1999)
`
`Fox and Bela (2000)
`Maidtar and
`
`Cameotra (2002)
`Yoneda et a1, (2005)
`Yeh et al. (2005)
`Amani et a1. (2010)
`
`Source: Adapted from Shaligram, NS. and Singhal, R.S. Food Tbchnol. Biotechnol.. 48: 119—134. 2010.
`
`HHIIILIJK
`
`091‘ I I I H I It‘ll. [I] U. mun Ii-
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 8 of 35
`
`

`

`LIPOPEPTIDE PRODUCTION REACTOR DESlGN
`AND OPTIMIZATiON
`
`Free and immobilized cells of B. .mbti'lis ATCC 21332 were grown to produce
`surfactin and fengycin (Chtioui et al., 2010). Although the production of both
`biosurfactants was enhanced by two to four times, fengycin was particularly
`improved. N-heptane was used for extracting the biosurfactant. A continuous
`extraction with a liquid membrane called petraction was used, but the stripping
`was too slow. Further optimization is needed. Petraction was also employed by
`Dimitrov et al. (60) for surfactin. At pH 5.65, 97% recovery was achieved com-
`pared to 83% at pH 6.05 in 4 h. However, approximately 90% was removed in
`30 min.
`
`Further studies were performed using a rotating disk bioreactor (Chtioui et a1.,
`2012). Cells were immobilized on the rotating disks. Foaming did not occur
`as the aeration was bubbleless. Fengycin production was favored (838 mg/L)
`compared to surfactin (212 mg/L). Increasing the number of disks improved
`the production of both products. Surfactin production was more correlated with
`improved oxygenation, while the fengycin production was related to more bio-
`film formation.
`
`1154
`
`Biosurfactants: Research Trends and Applications
`
`Various agro and industrial solid waste substrates including molasses, olive oil, and
`acrylarnide were evaluated. The biosurfactant was stable over the pH range of 5—9,
`and up to 5% NaCl and a temperature of 121°C. The surface tension was 28.6 mN/m.
`The lipopeptide was characterized as an octadecanoic acid methyl ester with four
`amino acids profileuigly-gly. This lipopeptide, thus, could have potential for micro-
`bial enhanced oil recovery and oil spill remediation.
`
`and aeration of 0.75 vvm. Primary inoculum age (55-57 h) of 5%—6% by
`
`A two-phase reactor with polyethylene glycol and dextran (D-40) was evaluated for
`surfactin production by B. subtilis ATCC 21332 (Drouin and Cooper, 1992) in a cyclone
`reactor. Cells accumulated in the dextran phase and surfactin in the other phase. This
`enabled the separation of surfactin from the cells to decrease cell inhibition.
`An airlift reactor in batch mode was employed to enhance aeration with a potato
`process effluent as the substrate (Noah et al., 2002). A 0.5 vvm air flow rate enabled
`surfactin removal. Conditions of a large inoculum, pH control, and the use of a
`pressurized reactor optimized the growth of B.
`.mbtilis over indigenous bacteria.
`Noah et a1. (2005) subsequently used a chemostat and low solid potato effluents.
`At 0.5 vvm, a surfactin concentration increased to 1.1 g/L was obtained at high agita—
`tion rates (400 rpm).
`Martinov et a1. (2008) studied aeration in a stirred tank reactor with foaming.
`Different agitators were tested due to the decrease in aeration in the presence of sur-
`factin. A low shear impeller Narcissus maintained stable kLa values while reducing
`foaming. Studies by Yeh et a1. (2006) indicated however that agitation rates above
`350 rpm and aeration above 2 vvrn lead to higher foaming levels that caused low
`surfactin production and low of cells. A he of 0.012/5 was optimal.
`Sen and Swaminathan (1997, 2004) studied surfactin production by B. subtilis
`3256. Maximal production (1.1 g/L) was at 314°C, pH 6.75. agitation of 140 rpm.
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 9 of 35
`
`

`

`Characterization, Production, and Applications of Lipopeptides
`
`155
`
`MEASUREMENT AND CHARACTERIZATION TECHNIQUES
`
`Enhanced surfactin production can be determined by blood agar plate screening
`due to hemolysis by surfactin (Mulligan et al., 1984). To verify that the isolates are
`biosurfactant producers, then the cultures must be grown and the surfactin levels
`determined. The most common technique for determining surfactant concentration
`is surface tension measurement and CMC determination. HPLC is also frequently
`used. An assay based on hemolysis was used for the analysis of surfactin in the
`fermentation broth. It was determined that the method could be used as a quick low-
`
`volume and secondary inoculation of (4—6 h) 9.5% by volume were also impor-
`tant for optimizing surfactin production.
`Gancel et a1. (2009) investigated lipopeptide production during cell immobili-
`zation on iron-enriched polypropylene particles. Immobilization improved biosurv
`factant production by up to 4.3 times. The amount of fengycin to surfactin varied
`depending on the iron content of the pellets. Highest surfactin (390 mg/L) and fengy-
`cin (680 mgfL) production was at 0.35% iron.
`Guez et a1. (2008) evaluated the influence of oxygen transfer rate on the produc«
`tion of the Iipopeptide mycolysin by B. subrilis ATCC6633. A respiratory activity
`monitoring system used for the study showed that oxygen metabolism has an effect
`on the homologue production and that the regulatory system is complex. Chenikher
`et al. (2010) examined the ability to control the specific growth rate for the produc-
`tion of surfactin and mycosubtilin. Most feeding strategies do not take into account
`the loss of the biomass with the foam. This must be taken into account to enable the
`maintenance of the specific growth rate and subsequently production. The growth
`rate of 0.05/11 was maintained.
`An integrated foam collector was integrated for biosurfactant production to study
`parameters for scale—up (Amani et al., 2010). The best conditions were 300 rpm and
`1.5 vvm for a surfactant yield on sucrose of 0.25 g/g. KLa of 0.011% was achievable in
`shake flasks and bioreactors, and this could potentially be used for scale-up.
`
`from 12 to 15 carbons. Leucine and isoleucine could be differentiated.
`
`technology method of surfactin analysis.
`Huang et al. (2009) compared blood plate hemolysis, surface tension, oil spread-
`ing, and demulsification. Surface tension measurement followed by demulsification
`tests allowed isolation of a demulsification strain Alcaligenes sp. S—Xj-l, which pro—
`duced a lipopeptide that was able to break O/W and W/O emulsions.
`,
`Knoblich et al. (1995) studied surfactin micelles by ice embedding and trans-
`mission electron cryomicroscopy. The micelles found were ellipsoidal with dimen—
`sions of 19, and 11 nm in width and length,respectively or spherical with a 5—9 nm
`in diameter, at pH 7. However, at pH 9.5, the micelles were more cylindrical with
`width and length dimensions of 10—14, and 40—160, or spherical with diameters of
`10—20 nm. Addition of 100 mM NaCl and 20 mM CaCl2 at pH 9.5 formed small
`
`spheres instead of the cylindrical micelles.
`Hue et al. (2001) examined the use of a combination of LSI-MS and MS/MS for
`the characterization of the mixtures of surfactin produced by B. subtih‘s. Amino acid
`composition was determined, and the length of the acyl chain was shown to vary
`
`lM‘illHlIk‘li- I
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 10 of 35
`
`

`

`156
`
`Biosurfactants: Research Trends and Applications
`
`Biosurfactant proteins produced by Lactobact‘llus fermentum RC—I4 have also
`been identified by a ProteinChip-interfaced mass spectrometer (Reid et al., 2002).
`Five tryptic peptide sequences by collision—induced dissociation tandem mass spec—
`trometry were identified following on-chip digestion of collagen—binding proteins.
`This may lead to the determination of the factors that are responsible for antistaphy~
`lococcal activity.
`lH—NMR was used by Bonmatin et a1. (1994) to show that surfactin can have two
`conformations depending on the pH. The saddle-like structure is bidentate with the
`two charged amino acids as sites for cation binding.
`SANS studies Were performed to study the characteristics of surfactin (Shen
`et al., 2009). At pH 7.5, the aggregation number was only 20, and the diameter of the
`micelles was 50 A with a hydrophobic core of 22 A radius. It is postulated that the
`leucines are in the hydrophobic core, which is consistent with its foaming characten
`istics. Further work (Shen et al., 2010) showed the solubilization of diphenylcarba~
`myl chloride.
`Pecci et al. (2010) characterized the biosurfactants produced by B. licheniformis
`V9T14 strain. This strain exhibited antimicrobial activity that inhibited biofilm for—
`mation of human pathogens. LC-ESI—MS/MS analyses were used, and fengycin and
`surfactin homologues were determined. Fractionation was further performed by
`silica gel chromatography. C13, C14. and C15 surfactin homologues were found plus
`C17 fengycins A and B. Other Cl4eC16 fengycin homologues were also confirmed.
`Most of the surfactin (61.3%) was in the C15 form with an MW of 1035. The two
`most common forms of fengycin A and B, respectively, were the C17 of MW 1477
`(25.1%) and 1505 (55.1%). The LC-ESIeMS/MS proved useful for the characteriza—
`tion of the lipopeptides.
`An oil emulsification test was used to screen for biosurfactants and bioemulsi-
`fiers for strains from a sea mud (Liu et al., 2010). A B. velezensr's H3 strain was
`isolated and could produce biosurfactants on starch and ammonium sulfate. C14 and
`C15 surfactins were discovered, which could lower the surface tension to 25.7 and
`27.0 mN/m, respectively, from pH 4 to 10. CMCs were in the order of 10'5 Incl/L.
`Antimicrobial properties were shown. The yield however was only 0.49 g/L. Highest
`yields of up to 50 g/L have been previously found by a strain on maltose and soybean
`flour (Yoneda et al., 2006).
`
`Pseudomonas sp. M1838. Arthrofactin are cyclic lipopeptides that function as
`
`GENETICS OF UPOPEPTIDE PRODUCTION
`
`Ultraviolet radiation mutation betWeen argC4 and hisAl on the genetic map led
`to a strain that produced 3.5 times surfactin (Mulligan et al., 1989). Another tech-
`nique included random mutagenesis by N—methyLN’ nitro-N-nitrosoguanidine of
`B. lichem'formis, where an increase in surfactin production of 12—fold was obtained
`(Lin et al., 1998). Tsuge et al. (2001) not only found that the yerP gene is involved in
`surfactin resistance in the strain but also evaluated if this gene was involved in sun
`factin production. Although the sfp gene was inserted into the strain. production was
`low. Therefore, it did not appear that the yer-P gene was linked to surfactin production.
`Washio et al.
`(2010) analyzed the genetics of arthrofactin production by
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 11 of 35
`
`

`

`Chelardi et al. (2012) also studied the motility of B. subrilis. They confirmed that
`.ver gene is needed for swarming, and surfactin increases surface wettability to
`allow swarming on low humidity surfaces.
`Various oil reservoirs of salinities from 2.1% to 15.9% were examined to deter-
`
`mine the presence of biosurfactant-producing strains (Simpson et al., 2011). The
`presence of surfactin (srfAj‘) and lichenysin (iicAj’) genes to evaluate the potential
`for biosurfactant production potential was confirmed. Subsequently, nutrient addi—
`tion was performed to stimulate production. This confirmed the ability to biostimu-
`lute biosurfactant production in an oil reservoir for oil recovery.
`
`EXTRACTION OF LIPOPEPTIDES
`
`Characterization, Production, and Applications of Lipopeptides
`
`157
`
`antibiotics, immunosuppressants, antitumor agents, siderophores, and surfactants.
`Schwartzer et a1. (2003) postulated it to be superior as a biosurfactant to surfactin
`and is necessary for the swarming and biofiim formation by the bacteria. Mutants
`from gene insertion were isolated that did not produce the biosurfactant, gaining
`some info on the synthesis.
`Ion beam implantation has also been utilized for generating high surfactine
`producing mutant of B. subrilis (Liu et al., 2006). N* is implanted by this method.
`Gong et a1. (2009) indicated that a mutant using this technique on the concentra-
`tion of a crude surfactin of 12.2 g/L could be produced from 6.5 g/L of biomass.
`It is not known what the effect of the implantation has on the metabolism of the
`microorganisms.
`
`Makkar and Cameotra (2001) also used foarn fractionation to rec0ver surfactin
`
`(‘rude extraction of lipopeptides is summarized in Table 6.3 but is usually by
`chemical extraction. For example, surfactin can be extracted by acid precipita~
`tion (pH 2) followed by solvent extraction by methanol (Vater et al., 2002). For
`Pxendomonns lipopeptides, multiple extraction by the ethyl acetate can be used
`{Kuiper et al., 2004).
`Purification of tipopeptides is important for subsequent industrial application. Thin
`layer chromatography, HPLC, gel permeation chromatography, ion exchange chroma—
`tography, and ultrafiltration have been used. HPLC by reverse phase chromatography
`in particular is often used with the detection by UV absorbance or mass spectrometry
`to provrde some information on the molecular mass of the components. Ultrafiltration
`with 30 kDa (UP-1) and 10 kDa (UF—II) cutoff membranes was employed in a single
`Step (Sivapathasekaran et al., 2011). The recovery was higher with the 10 cha mem_
`home (89%) compared to 73% with the 30 kDa. Purity was also higher (83% come
`Pared to 78%). The product was a mixture of surfactin and fengycin.
`The foaming characteristic of surfactin can be used to remove it during fer~
`mentation (Figure 6.4). At a concentration of 0.05 trig/L,
`it is comparable to
`SDS and bovine serum albumin (Razafindralambo et al., 1996). Low agitation
`3Foods in the fermentor were beneficial for the removal of high concentrations of
`surfactin in the collected foam (Davis et al., 2001). Between 10 and 30 h, stirrer
`SPEBdS of i46 and 166 rpm ted to surfactin concentrations of 1.67 and 1.22 g/L,
`re5-‘133Ctivc.=:ly, and agitation rates of 269 rpm produced concentrations of only
`75 mg/L. Overall recovery of the produced surfactin in the foam was over 90%.
`
`klilfh_ fl" 'IliiI-HKOIKIIU-m'n‘i
`
`PETITIONERS
`
`EXHIBIT NO. 1019 Page 12 of 35
`
`

`

`tion (Montastruc et at, 2008). Adsorption capacities were about 30 mg of surfactin
`
`by B. sabrilis MTCC 2423. Using sucrose as the substrate, yields of 4.5 g/L were
`obtained at 45°C. Other studies are shown in Table 6.4.
`Liu et a1. (2007) evaluated different conditions for adsorption for surfactin on
`activated carbon. pH values from 6.5 to 8.5 and 30°C were optimal. Adsorption
`onto activated carbon was studied to incorporate surfactin removal with produc-
`
`Source: Modified from Winterburn, LB. and Martin. R]. Biotecitnol. Letters, 34(2): 187—l 95.
`
`a Recovery = (CfVIIClV’i). where Cr, Ci are the foam and initial surfactin concentrations and V}. V[ are the
`initial liquid and foam volumes.
`
`5 Enrichment: Cfl'Cv
`M__w
`
`Air exhaust line
`
`Air outlet
`
`_
`Biosurfactant
`production
`
`Foam collection
`
`FIGURE 6.4 Recovery of surfactin by foam collection.
`
`Biosurfactants: Research Trends and Applications
`
`W T
`
`ABLE 6.3
`
`Recovery of Surfactin by Foam Collection
`Producing
`Foam
`
`Enrichment
`
`Generation
`Not controlled
`
`Recovery "/u “
`92.3
`
`Factorb
`55.0
`
`Not controlled
`
`28.7
`
`Controlled
`
`.
`
`50.1
`
`2.9
`
`Referen co
`Chen et al.
`(2006a)
`Chen et al.
`(2006b)
`Davis ct a1.
`(200!)
`
`Controlled
`
`Not controlled
`
`Organism
`B. subrilis
`BBK 006
`B. snbrilis
`BBK 006
`B. subtilis
`ATCC
`2133]
`
`Description
`Recovery during batch
`operation
`Recovery during
`continuous operation
`Separate foam
`fractionat

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket