Next Article in Journal
Thermotolerant Probiotic—The Potential of Improving the Survivability of Beneficial Bacteria
Previous Article in Journal
Effects of Lactobacillus plantarum-81-Fermented Feed on Growth and Intestinal Health of Muscovy Ducks
Previous Article in Special Issue
Characterization of Low pH and Inhibitor Tolerance Capacity of Candida krusei Strains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biofuel–Pharmaceutical Co-Production in Integrated Biorefineries: Strategies, Challenges, and Sustainability

1
Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Basic Medical, Guangdong Pharmaceutical University, Guangzhou 510006, China
2
School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
3
Key Laboratory of Renewable Energy, Guangzhou Institute of Energy Conversion, Chinese Academy of Sciences, Guangzhou 510640, China
*
Authors to whom correspondence should be addressed.
Fermentation 2025, 11(6), 312; https://doi.org/10.3390/fermentation11060312
Submission received: 4 April 2025 / Revised: 27 May 2025 / Accepted: 29 May 2025 / Published: 30 May 2025
(This article belongs to the Special Issue Biofuels and Green Technology)

Abstract

Global demands for sustainable energy and advanced therapeutics necessitate innovative interdisciplinary solutions. Integrated biorefining emerges as a strategic response, enabling the co-production of biofuels and pharmaceutical compounds through biomass valorization. This integrated model holds promise in enhancing resource utilization efficiency while ensuring economic viability. Our critical review methodically evaluates seven pivotal methodologies: seven key strategies: microbial metabolites, synthetic biology platforms, biorefinery waste extraction, nanocatalysts, computer-aided design, extremophiles, and plant secondary metabolites. Through systematic integration of these approaches, we reveal pivotal synergies and potential technological innovations that can propel multi-product biorefinery systems. Persistent challenges, particularly in reconciling complex metabolic flux balancing with regulatory compliance requirements, are analyzed. Nevertheless, advancements in systems biology, next-generation bioprocess engineering, and artificial intelligence-enhanced computational modeling present viable pathways for overcoming these obstacles. This comprehensive analysis substantiates the transformative capacity of integrated biorefining in establishing a circular bioeconomy framework, while underscoring the imperative of transdisciplinary cooperation to address existing technical and policy constraints.

1. Introduction

The 21st century faces two major global challenges: shifting to sustainable energy systems to address climate change and ensure energy security and continually searching for new and effective medicines to fight human diseases [1,2]. Although traditionally separate fields, energy production and pharmaceutical development are increasingly coming together, driven by advances in biotechnology and integrated biorefining [3,4,5]. The biorefinery concept, which involves processing biomass into a range of products like fuels, chemicals, and materials, provides a foundation for this synergy [6,7,8]. Strategically producing high-volume, lower value biofuels together with low-volume, high-value pharmaceuticals or their precursors and using shared feedstocks and infrastructure can significantly enhance the overall economic viability and sustainability of bio-based production [9,10,11]. Central to this integrated approach are microorganisms and plants; their vast metabolic diversity and suitability for engineering make them versatile biological factories [12,13].
With increasing interest in biorefineries and bio-based product manufacturing, there is an urgent need to systematically review advances in the integrated co-production of biofuels and pharmaceuticals. Furthermore, biorefineries can simultaneously produce other high-value substances (such as biochemicals including C1 compounds like carbon monoxide and hydrogen, C2 compounds such as ethanol, C3 compounds like glycerol and lactic acid, C4 compounds such as succinic acid, C5 derivatives like levulinic acid, and C6 derivatives such as 5-hydroxymethylfurfural and 2,5-furandicarboxylic acid), bioplastics (e.g., starch-based or lignin-based polymers), as well as proteins and fertilizers [14]. This review addresses that gap by providing a comprehensive overview of the strategies and technologies that enable the simultaneous production of these two valuable product streams within the same biorefinery framework. We examine a range of approaches, including the utilization of diverse microbial metabolites, the application of synthetic biology for targeted production, the valorization of biorefinery waste streams, the role of advanced nanocatalysts, the potential of extremophiles, and the integrated use of plant secondary metabolites (Figure 1). Additionally, we identify key challenges that must be overcome to make co-production economically and technically viable, such as optimizing metabolic pathways, developing efficient separation techniques, and scaling up processes. Furthermore, we clearly identify and discuss the overarching technical, economic, and regulatory challenges inherent in this interdisciplinary field, concluding with an outlook on future prospects and necessary advancements to fully realize its potential.
To ensure a comprehensive review, we conducted a systematic search across multiple databases, including PubMed, Scopus, Web of Science, and Google Scholar, to capture a wide range of relevant scientific literature. The search was performed using a combination of keywords such as biofuel, pharmaceutical, biorefinery, synthetic biology, metabolic engineering, nanocatalysts, extremophiles, and specific terms related to pharmaceutical compounds. The time range spanned from January 2010 to April 2025, with a particular emphasis on studies published between 2020 and 2025 to prioritize the most recent advancements in the field. Inclusion criteria were applied to select studies that addressed the integration of biofuel and pharmaceutical production within biorefineries or cross-disciplinary reference value, while exclusion criteria eliminated studies focusing solely on either biofuel or pharmaceutical production without their integration or capability. This review intentionally adopts a macro-level synthesis focusing on strategy interoperability and conceptual innovation rather than replicating process details already documented in references.

2. Dual-Purpose Development of Microbial Metabolites

Utilizing microbial metabolites provides a synergistic approach to achieving biofuel–pharmaceutical co-production. Microorganisms such as algae, bacteria, yeasts, and fungi possess diverse metabolic capabilities, enabling them to convert various feedstocks into biofuels like ethanol, butanol, lipids for biodiesel, or hydrogen, as well as high-value pharmaceutical compounds including antioxidants, anticancer agents, anti-inflammatory substances, pigments, and essential fatty acids [15]. This integrated strategy, often within biorefineries, aims to improve economic viability and sustainability. It does this by better utilizing biomass, sharing resources between production processes, possibly lowering separation and purification costs, and adding value to all major products from the microbial biomass [16]. Key microbial examples and their dual-output capabilities are systematically summarized in Table 1.
Selecting suitable microorganisms primarily hinges on the desired end products, the type of feedstock available—ranging from simple sugars to complex lignocellulosic materials or even CO2—and their compatibility with downstream processing requirements for both fuel and pharmaceutical-grade outputs. The microalga Haematococcus pluvialis is one of the most studied examples. This microorganism is renowned for accumulating astaxanthin, a potent antioxidant, at high concentrations—reaching up to 5% of its dry weight under stress conditions [30]. This carotenoid is widely used in nutraceuticals and studied for pharmaceutical uses because of its anti-inflammatory and neuroprotective properties. The residual algal biomass left after astaxanthin extraction contains valuable lipids and carbohydrates, making it an ideal feedstock for biodiesel production through transesterification, bioethanol via fermentation, or biogas through anaerobic digestion. This shows a clear path for sequential co-production [60]. Clostridium acetobutylicum is a key bacterium in the traditional Acetone-Butanol-Ethanol fermentation. It mainly produces butanol, a promising advanced biofuel [25]. However, its metabolism also produces butyric acid, an intermediate attracting interest for its potential anticancer properties, especially when used in targeted prodrugs like pivaloyloxymethyl butyrate [61,62]. The oleaginous yeast Phaffia rhodozyma naturally synthesizes astaxanthin while also accumulating significant quantities of lipids, particularly triglycerides, making it suitable for concurrent production of this valuable pigment and biodiesel precursors [63]. Similarly, other yeasts belonging to the Rhodotorula genus, such as Rhodotorula glutinis, are known co-producers, yielding lipids suitable for biodiesel alongside various carotenoids including β-carotene, torulene, and torularhodin, which function as antioxidants and potential provitamin A sources [64]. Specific strains like Rhodotorula babjevae Y-SL7 demonstrate remarkable capability on crude glycerol—a biodiesel byproduct, accumulating over 40% lipids while secreting potentially valuable polyol esters of fatty acids. Rhodotorula kratochvilovae SY89 further demonstrates this metabolic diversity through simultaneous production of lipids with yields reaching 66.7% of its biomass, along with carotenoids, immunostimulatory cell wall β-glucans, and extracellular glycolipids. [64,65]. The cyanobacterium Spirulina (genus Arthrospira, e.g., A. platensis), cultivated globally as a food supplement, produces the blue pigment phycocyanin, valued for its antioxidant, anti-inflammatory, and potential anticancer properties, alongside lipids that can be utilized for biodiesel production, showcasing co-production in photosynthetic microbes [66,67]. Oleaginous fungi, like Mortierella isabellina, accumulate lipids often rich in valuable polyunsaturated fatty acids such as gamma-linolenic acid, used therapeutically for inflammatory conditions, while the bulk lipid fraction remains suitable for biodiesel conversion [32]. Model organisms like the green alga Chlamydomonas reinhardtii offer potential for producing hydrogen or lipids as biofuels [68], coupled with β-carotene and potentially astaxanthin under specific stress conditions [23]. The extremophilic alga Dunaliella salina, which thrives in hypersaline environments, is commercially valued not only for its remarkably high β-carotene content reaching up to 14% of dry weight but also for accumulating lipids suitable for biodiesel production [26]. The versatile oleaginous yeast Yarrowia lipolytica has become a workhorse in metabolic engineering, capable of high lipid accumulation from various substrates and engineered to produce diverse compounds including omega-3 fatty acids, carotenoids, and even non-native products like flavonoids through synthetic biology approaches [69,70]. Bacterial systems, including species of Rhodococcus, and others like Bacillus, and Acinetobacter, known for lipid accumulation, can also be engineered or utilized for producing precursors like bioacetoin [71]. Other documented examples include the cyanobacterium Phormidium sp., which generates both lipids and squalene—a high value precursor for cosmetic and pharmaceutical applications [72,73], and the fungus Rhizopus oryzae generating lipids alongside chitin/chitosan polymers with biomedical applications [74].
The wide metabolic diversity found in various microbial groups—including algae, yeasts, bacteria, fungi, and archaea—provides many opportunities for the integrated production of biofuels and valuable pharmaceutical or nutraceutical compounds [75]. Further exploration of microbial diversity [76], combined with progress in metabolic engineering, synthetic biology [77], systems biology tools, and the development of efficient, integrated bioprocessing methods [78], is essential. These advances are needed to overcome current challenges and realize the full industrial potential of these dual-purpose microbial systems for making both energy and health products at the same time.

3. Dual Applications of Synthetic Biology Platforms

Synthetic biology offers a powerful engineering framework. It uses standardized genetic parts, quantitative modeling, and rational design principles to build and optimize biological systems for new purposes, including the customized co-production of biofuels and pharmaceuticals [79]. Furthermore, integrating high-throughput screening methods, biosensors for monitoring pathways dynamically, and advanced gene editing tools like CRISPR-Cas9 significantly speeds up the design–build–test–learn cycle, facilitating rapid optimization of these dual-production systems [77,80,81]. Table 2 summarizes representative examples of synthetic biology platforms for dual applications.
The practical application of synthetic biology in developing dual-purpose platforms is demonstrated through numerous successful examples across various microbial hosts [79]. Strains have been engineered to produce biofuels such as fatty acid ethyl esters or short-chain alkanes by introducing relevant biosynthetic pathways, while simultaneously optimizing native pathways or incorporating heterologous modules for the synthesis of valuable isoprenoids like lycopene or amorphadiene, the sesquiterpene precursor to the crucial antimalarial drug artemisinin [116]. Similarly, Saccharomyces cerevisiae, favored for its robustness in industrial fermentations, has been a prime target for synthetic biology approaches [117]. Seminal work led to yeast strains capable of producing high titers of artemisinic acid through the complex multi-gene engineering of the mevalonate pathway and introduction of amorphadiene synthase and P450 monooxygenase enzymes; this artemisinic acid is then chemically converted to artemisinin [116]. These same engineered yeast platforms, or parallel strains, can be optimized for the production of biofuels like ethanol, isobutanol, or the jet fuel precursor farnesene, often utilizing lignocellulosic hydrolysates as feedstock [117]. Other notable yeast engineering achievements include simultaneous production of ethanol with resveratrol, a high-value antioxidant linked to red wine, or with precursors to human insulin [118]. Photosynthetic microorganisms, such as cyanobacteria like Synechocystis sp. PCC 6803, offer the unique advantage of utilizing atmospheric CO2 as a carbon source [119]. Synthetic biology tools have been employed to redirect carbon fixed through photosynthesis towards biofuels like isobutanol, ethanol, or fatty acids, while also enhancing the production of native pigments with pharmaceutical potential including β-carotene and phycocyanin, or introducing pathways for novel pharmaceutical precursors [119]. The non-conventional oleaginous yeast Yarrowia lipolytica is another rapidly emerging platform, amenable to synthetic biology manipulations for the hyper-accumulation of lipids alongside engineered production of high-value molecules like omega-3 fatty acids or carotenoids [120].
Continued advances in synthetic biology tools, combined with deeper systems-level understanding and effective process integration, will be crucial for realizing the commercial potential of these dual-purpose platforms and establishing them as impactful industrial applications.

4. Extraction of Medicinal Components from Biorefinery Waste

The transition towards a circular bioeconomy necessitates maximizing value creation from biomass resources. Biorefineries, designed to convert biomass into fuels and chemicals, typically generate substantial amounts of side streams. Examples include lignin from lignocellulosic processing, spent microbial biomass after fermentation or extraction, and residual liquids such as hemicellulose hydrolysates or fermentation broth. These streams are often considered low-value waste or used for basic applications like animal feed or energy generation through combustion. Yet, they frequently hold a rich source of untapped bioactive components with potential uses in medicine, nutraceuticals, or cosmetics [121], as summarized in Table 3 which catalogs representative medicinal components extractable from major biorefinery side streams alongside their applications.
Lignocellulosic biorefineries process feedstocks including wood, agricultural residues such as straw and stover, or energy grasses, generating lignin as a major recalcitrant byproduct that typically accounts for 15–30% of the biomass. The complex polyphenolic structure of lignin renders it a promising source of valuable aromatic chemicals through depolymerization. Researchers employ diverse chemical methods like oxidation and hydrogenolysis, along with biological approaches including enzymatic treatment, to degrade lignin into lower molecular weight phenolic compounds. These breakdown products encompass vanillin, which serves as both flavorant and antioxidant; syringaldehyde; ferulic acid functioning as antioxidant and precursor; and p-coumaric acid with its antioxidant and anti-inflammatory properties—all finding established or emerging applications in pharmaceutical and food industries [153]. Hemicellulose hydrolysates, another stream from lignocellulosic pretreatment, primarily contain C5 and C6 sugars but also harbor minor yet potentially valuable components like phenolic acids (released from ester linkages) and oligosaccharides, particularly xylooligosaccharides derived from xylan. XOS are recognized for their prebiotic properties, promoting beneficial gut microbiota, and also exhibit antioxidant activity [154]. Microalgal biorefineries focused on lipid extraction for biodiesel leave behind substantial amounts of spent algal biomass. This protein-rich residue can be hydrolyzed to produce bioactive peptides with demonstrated antioxidant, antihypertensive (e.g., ACE-inhibitory), or antimicrobial activities [155]. Moreover, the residual biomass may retain valuable components including immunostimulatory β-glucans, various residual pigments such as carotenoids and chlorophyll derivatives, essential minerals, and residual omega-3 fatty acids, contingent upon the initial extraction efficiency [155]. Fermentation residues from bioethanol or ABE production, such as distiller’s dried grains with solubles from corn ethanol or the broth itself, are rich in microbial biomass, residual enzymes, proteins, and unfermented components including phenolic compounds derived from the original grain feedstock. These streams are potential sources for extracting enzymes, antimicrobial peptides, or antioxidant phytochemicals [156]. Similarly, biorefineries that process fungal biomass, particularly species such as Rhizopus oryzae used for lipid production, yield residues abundant in chitin and chitosan. These biocompatible polymers possess diverse biomedical applications owing to their hemostatic and antimicrobial properties, making them valuable for drug delivery systems, wound dressings, and tissue engineering scaffolds [157].
This “waste-to-value” strategy not only boosts profitability and diversifies product portfolios, but critically, it also supports resource circularity, minimizes environmental impact, and supplies sustainable sources of high-value compounds. This reinforces the argument that integrated biorefining is a cornerstone of the future bioeconomy.

5. Cross-Disciplinary Applications of Nanocatalysts

The convergence of nanotechnology and catalysis offers transformative potential for enhancing the efficiency, selectivity, and sustainability of chemical and biological processes central to integrated biofuel and pharmaceutical co-production [158]. In the biorefinery setting, nanocatalysts can be applied at various stages—from biomass pretreatment and fractionation to converting platform chemicals into advanced biofuels and synthesizing complex pharmaceutical intermediates [159]. This versatility makes them powerful cross-disciplinary tools for enabling new and efficient co-production pathways. Table 4 summarizes representative nanocatalyst types along with their catalyzed processes and co-production relevance.
One prominent area is the development of nano-biocatalysts, where enzymes are immobilized onto nanostructured supports, often magnetic nanoparticles (MNPs), nanocellulose, graphene derivatives, or mesoporous silica. Lipases immobilized on MNPs demonstrate particularly high efficiency and reusability in biodiesel production through algal oil or waste cooking oil transesterification, where their performance is enhanced by convenient magnetic separation capabilities [189]. Importantly, these same immobilized lipases can be utilized for enantioselective reactions critical in pharmaceutical synthesis, such as the kinetic resolution of chiral alcohols or esters used as drug precursors [190]. Metal nanoparticles supported on high surface-area materials such as carbon nanotubes, metal oxides, and polymers represent another important category of extensively studied nanocatalysts. Supported palladium, platinum, ruthenium, or gold NPs exhibit high activity in hydrogenation, dehydrogenation, oxidation, and C-C coupling reactions. These reactions are crucial for upgrading biomass-derived platform chemicals, such as furfural and 5-hydroxymethylfurfural (HMF), obtained from C5 and C6 sugars [191]. Selective hydrogenation of furfural produces either furfuryl alcohol, which serves as a resin precursor, or tetrahydrofurfuryl alcohol, known as a green solvent. Further processing can convert these compounds into fuel additives such as valeric biofuels or C8-C12 alkanes. Simultaneously, these platform chemicals and their derivatives serve as versatile building blocks for synthesizing various pharmaceutical intermediates and fine chemicals [191]. Nanocatalysts are also playing a key role in lignin valorization. Specifically designed metal or metal oxide nanocatalysts are being developed to selectively cleave particular bonds such as β-O-4 ether linkages within the complex lignin polymer under relatively mild conditions. This targeted approach aims to generate well-defined aromatic monomers, primarily phenolics, which can function as valuable precursors. These monomers have dual applications, serving both as building blocks for high-performance polymer components potentially useful in advanced biofuels or materials and as sources for bioactive compounds including antioxidants and anti-inflammatory agents [192]. Photocatalytic nanomaterials, such as titanium dioxide and zinc oxide nanoparticles or nanocomposites, utilize light energy to drive redox reactions. Their applications in biorefineries include assisting biomass pretreatment, degrading recalcitrant organic pollutants in wastewater streams [193], and potentially enabling specific organic syntheses relevant to pharmaceuticals under ambient conditions. Moreover, researchers are developing multifunctional and tandem nanocatalysts that combine different active sites such as acidic and metallic components within a single nanostructure. This integrated design enables cascade reactions capable of directly transforming raw biomass components or intermediate compounds into targeted biofuel and pharmaceutical precursors through streamlined reaction sequences, thereby significantly improving process intensification [194].
Further progress in catalyst design, synthesis, characterization, and reactor engineering is essential for moving nanocatalysts from laboratory-scale potential to reliable industrial processes. Such advances will improve the sustainability and economic viability of multi-product biorefineries [192].

6. Synergistic Optimization via Computer-Aided Design

The complexity inherent in biological systems and integrated biorefinery processes requires advanced tools for analysis, prediction, and optimization, particularly when targeting the synergistic co-production of multiple products like biofuels and pharmaceuticals [195]. Computer-aided design (CAD) offers such a framework. It includes a wide range of computational modeling, simulation, and data analysis techniques to handle this complexity [196]. By creating mathematical models of metabolic networks, genetic circuits, enzyme kinetics, reactor dynamics, and downstream processes, CAD tools allow researchers to conduct in silico experiments. These tools help test hypotheses, identify bottlenecks, predict outcomes of genetic or environmental changes, and guide the rational design of improved strains and processes [197].
As systematically summarized in Table 5, a diverse toolkit of computational methodologies is actively employed to optimize various aspects of biofuel and pharmaceutical co-production. At the core of metabolic engineering efforts are Genome-Scale Metabolic Models (GEMs), which represent the entire known metabolic reaction network of an organism [198]. Coupled with constraint-based modeling techniques like Flux Balance Analysis, GEMs enable the prediction of metabolic flux distributions under pseudo-steady-state conditions by optimizing objective functions such as biomass growth or product synthesis, while adhering to stoichiometric and thermodynamic constraints. Flux Balance Analysis and its many computational extensions, including OptFlux, Flux Variability Analysis, Minimization of Metabolic Adjustment, and Regulatory Flux Balance Analysis, serve as powerful tools for determining optimal genetic modifications. These approaches systematically identify gene knockout, knockdown, or overexpression targets that effectively redirect carbon flux toward the co-production of valuable compounds, such as achieving balanced synthesis of lipids for biodiesel and isoprenoids for pharmaceutical applications in engineered microbial systems like yeast or E. coli [198,199]. Computational pathway design tools such as RetroPath, BNICE, and OptStoic play a crucial role in discovering and assessing novel biosynthetic routes. These algorithms can construct artificial pathways by combining enzymes from diverse organisms to synthesize target compounds that either do not occur naturally or require optimized reaction kinetics and thermodynamics. A particularly valuable application involves deliberately engineering pathways that promote efficient co-production through strategic utilization of shared metabolic intermediates [200,201]. At the system level, advanced process simulation platforms including Aspen Plus, SuperPro Designer, and CHEMCAD facilitate comprehensive modeling and optimization of integrated biorefineries. These tools enable detailed analysis of the complete production chain, from feedstock pretreatment and enzymatic hydrolysis through fermentation processes to the intricate downstream separation and purification sequences necessary for multi-product biorefinery operations. These simulations allow for techno-economic analysis and life cycle assessment early in the design phase, evaluating the economic viability, energy balance, and environmental footprint of different co-production scenarios and process configurations [8,202]. More recently, machine learning and artificial intelligence algorithms are finding increasing application [203]. Machine learning enables the development of predictive models by analyzing large, complex datasets obtained from high-throughput experiments, such as those establishing correlations between genetic variants or fermentation parameters and co-product yields. These computational approaches can optimize fermentation media and culture conditions, predict novel enzymatic activities from sequence information, and facilitate the design of advanced synthetic genetic circuits for dynamic metabolic control. Such intelligent systems could empower microbial cells to autonomously regulate the equilibrium between biofuel and pharmaceutical synthesis in response to intracellular or environmental stimuli [203].
Integrating computational approaches across the entire research and development process—from initial strain design to full-scale process simulation and economic analysis—can significantly speed up progress. This acceleration helps realize the major economic and sustainability benefits that integrated, multi-product biorefineries offer.

7. Co-Development of Extremophiles for Dual Applications

Extremophiles are organisms adapted to thrive in environments hostile to most life, such as those with extreme temperature variations represented by thermophiles and psychrophiles, pH extremes including acidophiles and alkaliphiles, high salinity environments inhabited by halophiles, elevated pressure conditions tolerated by piezophiles, or intense radiation. These organisms are a valuable bioresource for industrial biotechnology, including the integrated co-production of biofuels and pharmaceuticals [232]. Their unique adaptations often result from novel biochemical pathways, distinct biomolecules, and highly robust enzymes known as extremozymes. These extremozymes can remain active and stable under the harsh processing conditions often found in industry [233,234].
The potential of extremophiles for simultaneous biofuel and pharmaceutical production is exemplified by various groups. Table 6 systematically summarizes representative extremophile species with demonstrated dual production capabilities, aligning their biotechnological applications with corresponding biochemical adaptations. Thermophilic bacteria and archaea, thriving at temperatures above 60 °C, are particularly attractive for processing lignocellulosic biomass. Organisms like Clostridium thermocellum, Caldicellulosiruptor species, and Thermoanaerobacterium saccharolyticum can directly ferment cellulose and hemicellulose components into biofuels such as ethanol or hydrogen at high temperatures, often in a process known as consolidated bioprocessing (CBP) which combines enzyme production, hydrolysis, and fermentation in a single step [66]. This high-temperature operation reduces contamination risks and can improve reaction kinetics. Crucially, these thermophiles are sources of highly thermostable enzymes, including cellulases, xylanases, ligninases, and amylases, which are themselves valuable products for various industrial applications, including biomass degradation, food processing, detergents, and potentially even pharmaceutical synthesis requiring robust catalysts. Halophilic microorganisms, which tolerate salt concentrations as high as saturation, offer distinct opportunities. Halophilic microalgae like Dunaliella salina are commercially cultivated in hypersaline ponds for their massive accumulation of β-carotene while also producing glycerol and lipids that could be targeted for biodiesel production [235]. Halophilic archaea, such as Halobacterium salinarum or Haloferax mediterranei, produce unique C50 carotenoids like bacterioruberin, exhibiting potent antioxidant activity potentially exceeding that of β-carotene [236]. They also synthesize osmoprotectants like ectoine, hydroxyectoine, and glycine betaine to maintain osmotic balance; these compounds are highly valuable as protein stabilizers and moisturizers in cosmetics and are explored for therapeutic applications [237]. Some haloarchaea also accumulate polyhydroxyalkanoates, biodegradable polyesters that can be considered bioplastics or potentially modified for drug delivery, alongside their potential role in biomass-for-energy schemes [238]. Psychrophilic microbes, found in permanently cold environments, produce lipases, proteases, and amylases that exhibit high catalytic activity at low temperatures [234]. This is advantageous for processes where heating is undesirable, such as food modification and detergent additives, or energetically costly [239]. While direct biofuel production at low temperatures is less common, these microbes can accumulate lipids rich in polyunsaturated fatty acids like EPA and DHA, which are high-value nutraceuticals, representing a potential co-product alongside cold-active enzymes [240]. Acidophiles and alkaliphiles, thriving at extreme pH values, similarly possess unique enzymes (e.g., pH-stable proteases, amylases) and metabolic capabilities that could be harnessed for specific biotransformations relevant to both biofuel upgrading and pharmaceutical synthesis under non-neutral pH conditions, potentially simplifying process integration or enabling novel reaction chemistries.
For certain industrial applications, the natural robustness, unique metabolic pathways, and production of extremozymes and novel bioactive compounds give extremophiles significant advantages over conventional mesophilic organisms [232]. Challenges remain, particularly in optimizing cultivation, developing genetic tools for non-model extremophiles, and scaling up processes [271]. Nevertheless, the vast untapped biodiversity in extreme environments represents a rich resource for future discoveries [272].

8. Integrated Utilization of Plant Secondary Metabolites

Plants are a primary source of biomass for biofuel production, mainly from lignocellulose or plant oils. At the same time, they produce a vast array of secondary metabolites. These compounds are not directly involved in primary growth but are vital for the plant’s defense, signaling, and adaptation. Many of these secondary metabolites—such as alkaloids, terpenoids, phenolics, and glycosides—have strong biological activities and are the basis for numerous pharmaceuticals, nutraceuticals, flavors, and fragrances.
As systematically summarized in Table 7 with their corresponding biofuel pathways and secondary metabolites, the potential for integrated utilization is evident across various plant feedstocks, ranging from dedicated energy crops and agricultural residues to established medicinal plants and oilseed crops. Lignocellulosic biomass from agricultural residues (e.g., corn stover, wheat straw, rice husks) or forestry residues, primarily targeted for cellulosic ethanol or biogas production, is also a source of phenolic compounds [273]. During conventional pretreatment methods such as dilute acid treatment, alkaline processing, and steam explosion—all employed to disrupt plant cell wall structures for biofuel production—phenolic acids including ferulic acid and p-coumaric acid are liberated into hydrolysate streams. These compounds typically exist in esterified forms bound to hemicellulose or lignin components prior to pretreatment. These compounds possess antioxidant and anti-inflammatory properties and can potentially be recovered as co-products before or after sugar fermentation [146]. Dedicated energy crops, such as switchgrass (Panicum virgatum), miscanthus (Miscanthus × giganteus), or fast-growing trees like poplar (Populus spp.), selected for high biomass yield, also contain a profile of secondary metabolites (phenolics, flavonoids, terpenoids) that could potentially be extracted as value-added co-products, although research in this area is less developed than for residues or medicinal plants. Medicinal plants offer a clear case for integration; Artemisia annua, cultivated for the potent antimalarial sesquiterpene lactone artemisinin, generates significant amounts of residual lignocellulosic biomass after the primary drug extraction. Developing methods to efficiently convert this residue into biofuels would substantially improve the economics of artemisinin production and reduce waste. Similarly, the processing of Taxus species needles or bark for the widely used anticancer drug paclitaxel leaves behind large quantities of biomass residues that could potentially be valorized through biofuel conversion pathways. Oilseed crops, the primary source for biodiesel, provide another important example. After oil extraction from seeds like rapeseed, sunflower, or soybean, a protein-rich meal remains. This meal is not only valuable as animal feed but also contains bioactive secondary metabolites. Rapeseed meal contains significant amounts of phenolic compounds, particularly sinapic acid and its derivatives, known for antioxidant properties [274]. Soybean meal is rich in isoflavones, phytoestrogens extensively studied for their potential health benefits, including roles in cancer prevention and cardiovascular health [275]. Integrating the recovery of these bioactive compounds from the meal fraction before or alongside its use as feed could add significant value to the biodiesel production chain. Key technological challenges involve developing cost-effective, selective, and scalable extraction and purification methods (e.g., sequential extraction, solvent optimization, membrane filtration, chromatography) that are compatible with subsequent biofuel processing steps and maintain the integrity of both the secondary metabolites and the residual biomass [276].
A key principle demonstrated across diverse feedstocks is that synergistic benefits arise from a holistic biorefining approach [313]. Targeting both the structural carbohydrates or oils for biofuels and the diverse secondary metabolites for high-value applications significantly broadens the economic foundation for utilizing energy crops, agricultural and forestry residues, traditional medicinal plants, and oilseed crops alike.

9. Overarching Challenges and Future Prospects for Integrated Co-Production

Despite the significant potential outlined in previous sections, realizing the widespread industrial application of integrated biofuel–pharmaceutical co-production faces considerable hurdles across multiple domains. Successfully navigating these is key to unlocking the full benefits of this synergistic approach.
Technically, the core challenges lie in efficiently managing complex biological systems for dual outputs and integrating the downstream processing of multiple, often disparate products. Optimizing cellular metabolism to simultaneously achieve high yields for both low-volume, high-value pharmaceuticals and high-volume, lower value biofuels without significant trade-offs due to metabolic burden or precursor competition remains a central difficulty (Reference metabolic burden/yield challenge). Furthermore, developing cost-effective, integrated downstream processes capable of efficiently separating and purifying products with potentially very different physicochemical properties and required purity levels (fuel-grade vs. pharmaceutical-grade) is a major bottleneck requiring significant innovation [314].
Economically, the substantial capital investment needed for establishing complex integrated biorefineries, coupled with uncertainties in long-term feedstock supply and pricing, poses major barriers. Achieving overall process profitability hinges on demonstrating cost-competitiveness, particularly for the biofuel component against established fossil fuels, while simultaneously meeting the stringent quality and regulatory standards for the high-value pharmaceutical co-product [315]. Seufitelli et al. (2022) demonstrated an integrated poplar biorefinery that co-produced jet fuel, xylitol, and formic acid with superior economic performance [316]. At a 250 kton/year scale, co-product revenues enabled a competitive jet fuel price of USD 3.13/gallon (vs. Conventional E) [316]. The Swedish company Sekab utilizes its CelluAPP technology to produce bioethanol and chemicals for the pharmaceutical industry (such as acetaldehyde and ethyl acetate). Its demonstration plant has showcased the potential for processing complex biomass [317]. The Norwegian company Borregaard’s biorefinery produces bioethanol and lignin-based products (such as vanillin and microfibrillated cellulose), the latter of which are used in the pharmaceutical industry; its operations highlight the potential for co-production [318,319]. Biodiesel refineries can enhance economic viability by producing high-value chemicals like succinic acid, but this requires extensive optimization to offset high capital costs [320]. Companies such as Abengoa, DuPont, and M&G abandoned their cellulosic ethanol plants before 2018 due to economic challenges, reflecting the profitability hurdles in the biofuel sector [321].
From a regulatory and societal perspective, clear, science-based, and internationally consistent frameworks are needed for the assessment and deployment of genetically modified organisms, especially those developed using advanced synthetic biology tools. Building public trust and market acceptance for novel bio-based processes and products through transparency and engagement is also crucial [322,323].
Nevertheless, continued progress across several key areas offers promising avenues to overcome these obstacles. Advances in synthetic biology and metabolic engineering provide increasingly powerful tools for designing and controlling complex cellular pathways with greater precision and efficiency. Systems biology approaches, integrating multi-omics data with computational modeling, deepen our understanding of cellular processes, enabling more predictive design and optimization. Concurrently, innovations in bioprocess engineering, focusing on process intensification and novel, selective separation technologies, are vital for improving scalability and reducing costs [324,325]. Performing rigorous techno-economic and life-cycle assessments throughout development is essential to guide research towards genuinely sustainable and viable configurations. Finally, fostering strong interdisciplinary collaborations and establishing supportive, stable policy environments that incentivize sustainable bio-based production are fundamental requirements for accelerating the transition from laboratory potential to industrial reality. Addressing these multifaceted challenges through concerted innovation holds the key to fully realizing the significant societal benefits offered by integrated biofuel–pharmaceutical co-production.
A particularly critical regulatory challenge lies in pharmaceutical Good Manufacturing Practice (GMP) compliance throughout production processes to ensure product quality and safety. The integration of GMP-compliant processes within biorefinery operations for simultaneous industrial chemical and pharmaceutical manufacturing presents multifaceted obstacles: It necessitates establishing independent quality management systems and dedicated facilities, substantially increasing capital and operational expenditures. Implementation of full-process traceability from raw materials to finished products becomes imperative to guarantee batch-to-batch consistency in co-produced pharmaceuticals. Three primary risk categories demand rigorous control in co-production systems: (1) comprehensive characterization of co-products (e.g., microbial metabolites, plant secondary metabolites), requiring thorough analysis of process residuals from biofuel operations such as lignin derivatives and nanoscale catalysts; (2) stringent evaluation of engineered biologics (e.g., therapeutic proteins from extremophiles) for immunogenicity and stability; and (3) regulatory compliance for genetically modified organisms (e.g., CRISPR-edited microbial strains), mandating adherence to compliance with specific genetically modified organism (GMO) regulations and elimination of genotoxic contaminants. Given pharmaceutical development cycles spanning 10–15 years with substantial clinical trial investments, the economic viability of co-production models becomes acutely dependent on production costs and market demand, while demanding biorefinery operators’ capacity to anticipate evolving regulatory landscapes. Regulatory pathways diverge significantly across co-product categories: biological products (e.g., insulin precursors from engineered yeast, therapeutic antibodies from transgenic tobacco plants) require Biologics License Applications (BLAs) through multistage clinical trials under FDA’s Center for Biologics Evaluation and Research (CBER) or European Medicines Agency (EMA) oversight [326]. Co-products meeting biosimilar criteria may leverage EMA’s abbreviated clinical trial requirements under recent guidelines. Meanwhile, bio-based chemical intermediates serving as pharmaceutical precursors (e.g., butanediol, lactic acid) primarily fall under industrial chemical regulations, though final drug products must still comply with stringent pharmacopeial standards [327].

10. Conclusions

Integrated co-production of biofuels and pharmaceuticals is a highly promising, synergistic strategy that uses biological systems to sustainably address global energy and health needs. Key approaches discussed include exploiting versatile microbial metabolites; harnessing the power of synthetic biology to engineer custom production platforms; and valorizing biorefinery waste streams into medicinal compounds. Moreover, applying advanced nanocatalysts, utilizing the potential of extremophiles, integrating plant secondary metabolites, and using computer-aided design to optimize complex interactions further illustrate the field’s multifaceted nature. These multifaceted strategies are further contextualized in Figure 2, which suggests critical future research directions to advance this field. Significant technical, economic, and regulatory challenges remain. However, future directions based on systems biology, process intensification, robust strain development, and supportive policies are set to unlock co-production’s substantial potential. Through continued interdisciplinary innovation and collaboration, this field can significantly contribute to a circular bioeconomy, energy security, and the discovery of novel therapeutics.

Author Contributions

Conceptualization, H.Y. and W.L.; methodology, M.H.; validation, R.S.; formal analysis, T.L.; investigation, M.H.; resources, M.H.; data curation, T.L.; writing—original draft preparation, T.L. and W.L.; writing—review and editing, H.Y. and W.L.; visualization, M.H. and R.S.; supervision, W.L.; project administration, W.L.; funding acquisition, T.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Key Area Project of General Universities in Guangdong Province (No. 2024ZDZX2080, and the College Students’ Innovation and Entrepreneurship Training Program (No. 202310573025 and 202410573003X).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The authors would like to express their sincere gratitude to Jiayuan Zhou from Guangzhou University of Chinese Medicine for her valuable assistance in refining the language and structure of this manuscript. Her expertise in medical English significantly improved the clarity and coherence of our interdisciplinary research.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Moustakas, K.; Loizidou, M.; Rehan, M.; Nizami, A.S. A review of recent developments in renewable and sustainable energy systems: Key challenges and future perspective. Renew. Sustain. Energy Rev. 2020, 119, 109418. [Google Scholar] [CrossRef]
  2. Espro, C.; Paone, E.; Mauriello, F.; Gotti, R.; Uliassi, E.; Bolognesi, M.L.; Rodríguez-Padrón, D.; Luque, R. Sustainable production of pharmaceutical, nutraceutical and bioactive compounds from biomass and waste. Chem. Soc. Rev. 2021, 50, 11191–11207. [Google Scholar] [CrossRef] [PubMed]
  3. Abdullah, M.A.; Hussein, H.A. Hussein. Integrated algal and oil palm biorefinery as a model system for bioenergy co-generation with bioproducts and biopharmaceuticals. Bioresour. Bioprocess. 2021, 8, 40. [Google Scholar] [CrossRef]
  4. Adetunji, C.O.; Olaniyan, O.T.; Anani, O.A.; Bodunrinde, R.E.; Osemwegie, O.O.; Ubi, B.E. Chapter 17—Integrated processes for production of pharmaceutical products from agro-wastes. In Biomass, Biofuels, Biochemicals; Elsevier: Amsterdam, The Netherlands, 2022; pp. 439–461. [Google Scholar]
  5. Kavitha, S.; Ravi, Y.K.; Gunasekaran, M.; Chattopadhyay, I.; Palani, S.; Kumar, V.; Kumar, G.; Jeyakumar, R.B. Development of an Integrated Biorefinery System for Bioconversion of Lignocellulosic Biomass to Polyhydroxyalkanoates and Biohydrogen. ACS Sustain. Chem. Eng. 2023, 11, 4606–4622. [Google Scholar]
  6. Marzban, N.; Psarianos, M.; Herrmann, C.; Schulz-Nielsen, L.; Olszewska-Widdrat, A.; Arefi, A.; Pecenka, R.; Grundmann, P.; Schlüter, O.K.; Hoffmann, T.; et al. Smart integrated biorefineries in bioeconomy: A concept toward zero-waste, emission reduction, and self-sufficient energy production. Biofuel Res. J. 2025, 12, 2319–2349. [Google Scholar] [CrossRef]
  7. Tang, D.Y.Y.; Yew, G.Y.; Koyande, A.K.; Chew, K.W.; Vo, D.-V.N.; Show, P.L. Green technology for the industrial production of biofuels and bioproducts from microalgae: A review. Environ. Chem. Lett. 2020, 18, 1967–1985. [Google Scholar] [CrossRef]
  8. Pérez-Almada, D.; Galán-Martín, Á.; Contreras, M.d.M.; Castro, E. Integrated techno-economic and environmental assessment of biorefineries: Review and future research directions. Sustain. Energy Fuels 2023, 7, 4031–4050. [Google Scholar] [CrossRef]
  9. Koyande, A.K.; Show, P.L.; Guo, R.; Tang, B.; Ogino, C.; Chang, J.S. Bio-processing of algal bio-refinery: A review on current advances and future perspectives. Bioengineered 2019, 10, 574–592. [Google Scholar] [CrossRef]
  10. Sari, Y.W.; Kartikasari, K.; Widyarani; Setyaningsih, I.; Lestari, D. Chapter 13—Techno-economic assessment of microalgae for biofuel, chemical, and bioplastic. In Microalgae; Galanakis, C.M., Ed.; Academic Press: London, UK, 2021; pp. 409–432. [Google Scholar]
  11. Rosero-Chasoy, G.; Rodríguez-Jasso, R.M.; Aguilar, C.N.; Buitrón, G.; Chairez, I.; Ruiz, H.A. Microbial co-culturing strategies for the production high value compounds, a reliable framework towards sustainable biorefinery implementation—An overview. Bioresour. Technol. 2021, 321, 124458. [Google Scholar] [CrossRef]
  12. Granata, T.; Rattenbacher, B.; Kehl, F.; Egli, M. Microbial factories and exploiting synergies of bioreactor technologies to produce bioproducts. Fermentation 2024, 10, 135. [Google Scholar] [CrossRef]
  13. Kumar, R.; Ghosh, A.K.; Pal, P. Synergy of biofuel production with waste remediation along with value-added co-products recovery through microalgae cultivation: A review of membrane-integrated green approach. Sci. Total Environ. 2020, 698, 134169. [Google Scholar] [CrossRef] [PubMed]
  14. Calvo-Flores, F.G.; Martin-Martinez, F.J. Biorefineries: Achievements and challenges for a bio-based economy. Front. Chem. 2022, 10, 973417. [Google Scholar] [CrossRef]
  15. Costa, G.d.S.; Martinez-Burgos, W.J.; dos Reis, G.A.; Puche, Y.P.; Vega, F.R.; Rodrigues, C.; Serra, J.L.; Campos, S.d.M.; Soccol, C.R. Advances in biomass and microbial lipids production: Trends and prospects. Processes 2024, 12, 2903. [Google Scholar] [CrossRef]
  16. YousefiPour, M.; Gheibipour, M.; Yousefipour, M.; Gheibipour, F.; Dar, M.A. Chapter 25—Microbial production of biofuels: An insight into cocultivation, challenges, and future prospects. In Biofuels and Sustainability; Zhu, D., Dar, M.A., Shahnawaz, M., Eds.; Elsevier Science Ltd.: Amsterdam, The Netherlands, 2025; pp. 423–439. [Google Scholar]
  17. Tiku, V.; Kew, C.; Kofoed, E.M.; Peng, Y.; Dikic, I.; Tan, M.-W. Acinetobacter baumannii secretes a bioactive lipid that triggers inflammatory signaling and cell death. Front. Microbiol. 2022, 13, 870101. [Google Scholar] [CrossRef]
  18. Bjerk, T.R.; Severino, P.; Jain, S.; Marques, C.; Silva, A.M.; Pashirova, T.; Souto, E.B. Biosurfactants: Properties and applications in drug delivery, biotechnology and ecotoxicology. Bioengineering 2021, 8, 115. [Google Scholar] [CrossRef]
  19. Wang, A.; Meng, D.; Hao, Q.; Xia, R.; Zhang, Q.; Ran, C.; Yang, Y.; Li, D.; Liu, W.; Zhang, Z. Effect of supplementation of solid-state fermentation product of Bacillus subtilis HGcc-1 to high-fat diet on growth, hepatic lipid metabolism, epidermal mucus, gut and liver health and gut microbiota of zebrafish. Aquaculture 2022, 560, 738542. [Google Scholar] [CrossRef]
  20. Saini, M.; Anu; Rapoport, A.; Tiwari, S.K.; Singh, D.; Malik, V.; Kumar, S.; Singh, B. Bioacetoin production by Bacillus subtilis subsp. subtilis using enzymatic hydrolysate of lignocellulosic biomass. Fermentation 2023, 9, 698. [Google Scholar] [CrossRef]
  21. Hippler, M.; Khosravitabar, F. Light-Driven H2 Production in Chlamydomonas reinhardtii: Lessons from Engineering of Photosynthesis. Plants 2024, 13, 2114. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, Y.; Hu, Y.; Mo, J.; Wong, T.Y.; Liu, J.; Alessandro, P.; Tang, B.Z.; Wang, W.-X.; Yan, N. Bioprospecting of Chlamydomonas reinhardtii for boosting biofuel-related products production based on novel aggregation-induced emission active extracellular polymeric substances nanoprobes. Bioresour. Technol. 2024, 399, 130636. [Google Scholar] [CrossRef]
  23. Chen, Y.; Du, H.; Liang, H.; Hong, T.; Li, T. Enhanced Carotenoid Production in Chlamydomonas reinhardtii by Overexpression of Endogenousand Exogenous Beta-Carotene Ketolase (BKT) Genes. Int. J. Mol. Sci. 2023, 24, 11382. [Google Scholar] [CrossRef]
  24. Nailwal, V.; Mukherjee, T.; Mohan, S.V. Decoupling acidogenic cascade via bio-electrogenic induced ABE pathway for enhanced n-butanol synthesis in Clostridium acetobutylicum ATCC-824: Bioenergetics and gene expression analysis. Chem. Eng. J. 2024, 486, 150015. [Google Scholar] [CrossRef]
  25. Chang, W.L.; Hou, W.; Xu, M.; Yang, S.T. High-rate continuous n-butanol production by Clostridium acetobutylicum from glucose and butyric acid in a single-pass fibrous-bed bioreactor. Biotechnol. Bioeng. 2022, 119, 3474–3486. [Google Scholar] [CrossRef] [PubMed]
  26. Goswami, R.K.; Agrawal, K.; Verma, P. Microalgae Dunaliella as biofuel feedstock and β-carotene production: An influential step towards environmental sustainability. Energy Convers. Manag. X 2022, 13, 100154. [Google Scholar] [CrossRef]
  27. Ludwig, K.; Rihko-Struckmann, L.; Brinitzer, G.; Unkelbach, G.; Sundmacher, K. β-Carotene extraction from Dunaliella salina by supercritical CO2. J. Appl. Phycol. 2021, 33, 1435–1445. [Google Scholar] [CrossRef]
  28. Vitucci, D.; Amoresano, A.; Nunziato, M.; Muoio, S.; Alfieri, A.; Oriani, G.; Scalfi, L.; Frusciante, L.; Rigano, M.M.; Pucci, P.; et al. Nutritional Controlled Preparation and Administration of Different Tomato Purées Indicate Increase of β-Carotene and Lycopene Isoforms, and of Antioxidant Potential in Human Blood Bioavailability: A Pilot Study. Nutrients 2021, 13, 1336. [Google Scholar] [CrossRef]
  29. Hosseini, A.; Jazini, M.; Mahdieh, M.; Karimi, K. Efficient superantioxidant and biofuel production from microalga Haematococcus pluvialis via a biorefinery approach. Bioresour. Technol. 2020, 306, 123100. [Google Scholar] [CrossRef]
  30. Zarei, Z.; Zamani, H. Biorefinery approach to stimulate astaxanthin and biofuel generation in microalga Haematococcus pluvialis under different light irradiance. Clean Technol. Environ. Policy 2024, 26, 3333–3347. [Google Scholar] [CrossRef]
  31. Masoudi, A.; Jorjani, M.; Alizadeh, M.; Mirzamohammadi, S.; Mohammadi, M. Anti-inflammatory and antioxidant effects of astaxanthin following spinal cord injury in a rat animal model. Brain Res. Bull. 2021, 177, 324–331. [Google Scholar] [CrossRef]
  32. Sallet, D.; Ugalde, G.A.; Tres, M.V.; Mazutti, M.A.; Zabot, G.L.; Kuhn, R.C. Oil and Biodiesel Production from Mortierella isabellina Biomass by a Direct Near-Critical Fluid Extraction and Transesterification Method. Biomass 2025, 5, 6. [Google Scholar] [CrossRef]
  33. Papanikolaou, S.; Aggelis, G. Sources of microbial oils with emphasis to Mortierella (Umbelopsis) isabellina fungus. World J. Microbiol. Biotechnol. 2019, 35, 63. [Google Scholar] [CrossRef]
  34. Baker, E.J.; Valenzuela, C.A.; van Dooremalen, W.T.; Martínez-Fernández, L.; Yaqoob, P.; Miles, E.A.; Calder, P.C. Gamma-linolenic and pinolenic acids exert anti-inflammatory effects in cultured human endothelial cells through their elongation products. Mol. Nutr. Food Res. 2020, 64, 2000382. [Google Scholar] [CrossRef] [PubMed]
  35. Mussagy, C.U.; dos Santos, A.; Militão, G.F.; de Oliveira, J.C.S.; Umbuzeiro, G.d.A.; Peixoto, G.; Pessoa Jr, A.; Santos-Ebinuma, V.C. Phaffia rhodozyma biorefinery: A sustainable pathway to obtain natural pigments and production of methane biogas as renewable fuel. Chem. Eng. J. 2023, 473, 145350. [Google Scholar] [CrossRef]
  36. Lai, J.-X.; Chen, X.; Bu, J.; Hu, B.-B.; Zhu, M.-J. Direct production of astaxanthin from food waste by Phaffia rhodozyma. Process Biochem. 2022, 113, 224–233. [Google Scholar] [CrossRef]
  37. Jeong, Y.; Cho, S.H.; Lee, H.; Choi, H.K.; Kim, D.M.; Lee, C.G.; Cho, S.; Cho, B.K. Current Status and Future Strategies to Increase Secondary Metabolite Production from Cyanobacteria. Microorganisms 2020, 8, 1849. [Google Scholar] [CrossRef]
  38. Fagundes, M.B.; Alvarez-Rivera, G.; Vendruscolo, R.G.; Voss, M.; da Silva, P.A.; Barin, J.S.; Jacob-Lopes, E.; Zepka, L.Q.; Wagner, R. Green microsaponification-based method for gas chromatography determination of sterol and squalene in cyanobacterial biomass. Talanta 2021, 224, 121793. [Google Scholar] [CrossRef] [PubMed]
  39. Dash, A.; Banerjee, R. Exploring indigenously produced celite-immobilized Rhizopus oryzae NRRL 3562-lipase for biodiesel production. Energy 2021, 222, 119950. [Google Scholar] [CrossRef]
  40. Beliaeva, A.; Nianikova, G.; Rostovtseva, P. Chitin-chitosan complex from Rhizopus oryzae obtained on a pea culture medium, and some of its physicochemical properties. E3S Web Conf. 2020, 215, 06001. [Google Scholar] [CrossRef]
  41. El-Zawawy, N.A.; Ali, S.S.; Nouh, H.S. Exploring the potential of Rhizopus oryzae AUMC14899 as a novel endophytic fungus for the production of l-tyrosine and its biomedical applications. Microb. Cell Fact. 2023, 22, 31. [Google Scholar] [CrossRef]
  42. Guerfali, M.; Ayadi, I.; Ayadi, W.; Smaoui, S.; Elhadef, K.; Zaghden, H.; Jlaiel, L.; Sahli, E.; Belghith, H.; Gargouri, A. Concomitant production of multifunctional metabolites on biodiesel-derived crude glycerol by the oleaginous yeast Rhodotorula babjevae Y-SL7. Biomass Convers. Biorefin. 2024, 14, 10237–10250. [Google Scholar] [CrossRef]
  43. Garay, L.A.; Sitepu, I.R.; Cajka, T.; Cathcart, E.; Fiehn, O.; German, J.B.; Block, D.E.; Boundy-Mills, K.L. Simultaneous production of intracellular triacylglycerols and extracellular polyol esters of fatty acids by Rhodotorula babjevae and Rhodotorula aff. paludigena. J. Ind. Microbiol. Biotechnol. 2017, 44, 1397–1413. [Google Scholar] [CrossRef]
  44. Li, Z.; Li, C.; Cheng, P.; Yu, G. Rhodotorula mucilaginosa—Alternative sources of natural carotenoids, lipids, and enzymes for industrial use. Heliyon 2022, 88, e10888. [Google Scholar] [CrossRef] [PubMed]
  45. Vargas-Sinisterra, A.F.; Ramírez-Castrillón, M. Yeast carotenoids: Production and activity as antimicrobial biomolecule. Arch. Microbiol. 2021, 203, 873–888. [Google Scholar] [CrossRef] [PubMed]
  46. Byrtusová, D.; Szotkowski, M.; Kurowska, K.; Shapaval, V.; Márová, I. Rhodotorula kratochvilovae CCY 20-2-26—The source of multifunctional metabolites. Microorganisms 2021, 9, 1280. [Google Scholar] [CrossRef]
  47. Szotkowski, M.; Holub, J.; Šimanský, S.; Hubačová, K.; Sikorová, P.; Mariničová, V.; Němcová, A.; Márová, I. Bioreactor co-cultivation of high lipid and carotenoid producing yeast Rhodotorula kratochvilovae and several microalgae under stress. Microorganisms 2021, 9, 1160. [Google Scholar] [CrossRef]
  48. Wu, L.; Zhao, J.; Zhang, X.; Liu, S.; Zhao, C. Antitumor effect of soluble β-glucan as an immune stimulant. Int. J. Biol. Macromol. 2021, 179, 116–124. [Google Scholar] [CrossRef]
  49. Bhatia, S.K.; Gurav, R.; Choi, Y.-K.; Lee, H.-J.; Kim, S.H.; Suh, M.J.; Cho, J.Y.; Ham, S.; Lee, S.H.; Choi, K.-Y. Rhodococcus sp. YHY01 a microbial cell factory for the valorization of waste cooking oil into lipids a feedstock for biodiesel production. Fuel 2021, 301, 121070. [Google Scholar] [CrossRef]
  50. Maltseva, P.Y.; Plotnitskaya, N.A.; Ivshina, I.B. Transformation of Terpenoids and Steroids Using Actinomycetes of the Genus Rhodococcus. Molecules 2024, 29, 3378. [Google Scholar] [CrossRef]
  51. Zappaterra, F.; Costa, S.; Summa, D.; Bertolasi, V.; Semeraro, B.; Pedrini, P.; Buzzi, R.; Vertuani, S. Biotransformation of cortisone with Rhodococcus rhodnii: Synthesis of new steroids. Molecules 2021, 26, 1352. [Google Scholar] [CrossRef] [PubMed]
  52. Haghighi, M.; Zare, L.B.; Ghiasi, M. Biodiesel production from Spirulina algae oil over [Cu(H2PDC)(H2O)2] complex using transesterification reaction: Experimental study and DFT approach. Chem. Eng. J. 2022, 430, 132777. [Google Scholar] [CrossRef]
  53. Li, Y. The bioactivities of phycocyanobilin from spirulina. J. Immunol. Res. 2022, 2022, 4008991. [Google Scholar] [CrossRef]
  54. Naveira-Pazos, C.; Robles-Iglesias, R.; Fernández-Blanco, C.; Veiga, M.C.; Kennes, C. State-of-the-art in the accumulation of lipids and other bioproducts from sustainable sources by Yarrowia lipolytica. Rev. Environ. Sci. Bio/Technol. 2023, 22, 1131–1158. [Google Scholar] [CrossRef]
  55. Cao, L.; Yin, M.; Shi, T.-Q.; Lin, L.; Ledesma-Amaro, R.; Ji, X.-J. Engineering Yarrowia lipolytica to produce nutritional fatty acids: Current status and future perspectives. Synth. Syst. Biotechnol. 2022, 7, 1024–1033. [Google Scholar] [CrossRef]
  56. Liu, M.; Zhang, J.; Ye, J.; Qi, Q.; Hou, J. Morphological and metabolic engineering of Yarrowia lipolytica to increase β-carotene production. ACS Synth. Biol. 2021, 10, 3551–3560. [Google Scholar] [CrossRef] [PubMed]
  57. Lv, Y.; Marsafari, M.; Koffas, M.; Zhou, J.; Xu, P. Optimizing oleaginous yeast cell factories for flavonoids and hydroxylated flavonoids biosynthesis. ACS Synth. Biol. 2019, 8, 2514–2523. [Google Scholar] [CrossRef]
  58. Ru, Z.; Liu, M.; Chen, Q.; Li, H.; Ning, Y.; Zeng, W.; Zhou, J. High-Level De Novo Production of (2S)-Naringenin in Yarrowia lipolytica Using Metabolic and Enzyme Engineering. ACS Agric. Sci. Technol. 2025, 5, 784–793. [Google Scholar] [CrossRef]
  59. da Silva, J.L.; Sales, M.B.; de Castro Bizerra, V.; Nobre, M.M.R.; de Sousa Braz, A.K.; da Silva Sousa, P.; Cavalcante, A.L.; Melo, R.L.; Gonçalves De Sousa Junior, P.; Neto, F.S. Lipase from Yarrowia lipolytica: Prospects as an industrial biocatalyst for biotechnological applications. Fermentation 2023, 9, 581. [Google Scholar] [CrossRef]
  60. Oslan, S.N.H.; Shoparwe, N.F.; Yusoff, A.H.; Rahim, A.A.; Chang, C.S.; Tan, J.S.; Oslan, S.N.; Arumugam, K.; Ariff, A.B.; Sulaiman, A.Z.; et al. A Review on Haematococcus pluvialis Bioprocess Optimization of Green and Red Stage Culture Conditions for the Production of Natural Astaxanthin. Biomolecules 2021, 11, 256. [Google Scholar] [CrossRef]
  61. Lehmann, D.; Radomski, N.; Lütke-Eversloh, T. New insights into the butyric acid metabolism of Clostridium acetobutylicum. Appl. Microbiol. Biotechnol. 2012, 96, 1325–1339. [Google Scholar] [CrossRef]
  62. Dwidar, M.; Park, J.Y.; Mitchell, R.J.; Sang, B.I. The future of butyric acid in industry. Sci. World J. 2012, 2012, 471417. [Google Scholar] [CrossRef]
  63. Jia, J.; Chen, Z.; Li, Q.; Li, F.; Liu, S.; Bao, G. The enhancement of astaxanthin production in Phaffia rhodozyma through a synergistic melatonin treatment and zinc finger transcription factor gene overexpression. Front. Microbiol. 2024, 15, 1367084. [Google Scholar] [CrossRef]
  64. EElfeky, N.; Elmahmoudy, M.; Zhang, Y.; Guo, J.; Bao, Y. Lipid and Carotenoid Production by Rhodotorula glutinis with a Combined Cultivation Mode of Nitrogen, Sulfur, and Aluminium Stress. Fermentation 2019, 9, 2444. [Google Scholar] [CrossRef]
  65. Tang, W.; Wang, Y.; Zhang, J.; Cai, Y.; He, Z. Biosynthetic Pathway of Carotenoids in Rhodotorula and Strategies for Enhanced Their Production. J. Microbiol. Biotechnol. 2019, 29, 507–517. [Google Scholar] [CrossRef]
  66. Russo, N.; Ballotta, M.; Usai, L.; Torre, S.; Giordano, M.; Fais, G.; Casula, M.; Dessì, D.; Nieri, P.; Damergi, E.; et al. Mixotrophic Cultivation of Arthrospira platensis (Spirulina) under Salt Stress: Effect on Biomass Composition, FAME Profile and Phycocyanin Content. Mar. Drugs 2024, 22, 381. [Google Scholar] [CrossRef]
  67. Shayesteh, H.; Laird, D.W.; Hughes, L.J.; Nematollahi, M.A.; Kakhki, A.M.; Moheimani, N.R. Co-Producing Phycocyanin and Bioplastic in Arthrospira platensis Using Carbon-Rich Wastewater. BioTech 2023, 12, 49. [Google Scholar] [CrossRef] [PubMed]
  68. Scranton, M.A.; Ostrand, J.T.; Fields, F.J.; Mayfield, S.P. Chlamydomonas as a model for biofuels and bio-products production. Plant J. 2015, 82, 523–531. [Google Scholar] [CrossRef]
  69. Park, Y.K.; Nicaud, J.M. Metabolic Engineering for Unusual Lipid Production in Yarrowia lipolytica. Microorganisms 2020, 8, 1937. [Google Scholar] [CrossRef] [PubMed]
  70. Gu, Y.; Lu, X.; Liu, T.; Song, Y.; Sang, E.; Ding, S.; Liu, L.; Xue, F.; Xu, P. Engineering the oleaginous yeast Yarrowia lipolytica to produce nutraceuticals: From metabolic design to industrial applications. Food Bioeng. 2023, 2, 62–73. [Google Scholar] [CrossRef]
  71. Shields-Menard, S.; Amirsadeghi, M.; Sukhbaatar, B.; Revellame, E.; Hernandez, R.; Donaldson, J.; French, T. Lipid accumulation by Rhodococcus rhodochrous grown on glucose. J. Ind. Microbiol. Biotechnol. 2015, 42, 693–699. [Google Scholar] [CrossRef]
  72. Shalu, S.; Karthikanath, P.K.R.; Vaidyanathan, V.K.; Blank, L.M.; Germer, A.; Balakumaran, P.A. Microbial Squalene: A Sustainable Alternative for the Cosmetics and Pharmaceutical Industry—A Review. Life Sci. 2024, 24, e202400003. [Google Scholar] [CrossRef]
  73. Koch, M.; Noonan, A.J.C.; Qiu, Y.; Dofher, K.; Kieft, B.; Mottahedeh, S.; Shastri, M.; Hallam, S.J. The survivor strain: Isolation and characterization of Phormidium yuhuli AB48, a filamentous phototactic cyanobacterium with biotechnological potential. Front. Bioeng. Biotechnol. 2022, 10, 932695. [Google Scholar] [CrossRef]
  74. Satari, B.; Karimi, K.; Taherzadeh, M.J.; Zamani, A. Co-Production of Fungal Biomass Derived Constituents and Ethanol from Citrus Wastes Free Sugars without Auxiliary Nutrients in Airlift Bioreactor. Fermentation 2016, 17, 302. [Google Scholar] [CrossRef] [PubMed]
  75. Martien, J.I.; Amador-Noguez, D. Recent applications of metabolomics to advance microbial biofuel production. Curr. Opin. Biotechnol. 2017, 43, 118–126. [Google Scholar] [CrossRef] [PubMed]
  76. Satheesh, S.; Cherian, T.; Varghese, T.; Eranhottu, S.; Shahulhameed, F.P.P. Bioprospecting of Microorganisms for Biofuel Production: Metabolic Engineering, Applications, and Challenges. In Catalytic Applications of Biochar for Environmental Remediation: Valorization of Lignocellulosic Waste Biomass into Bioenergy; American Chemical Society: Washington, DC, USA, 2024; Volume 3, pp. 91–107. [Google Scholar]
  77. Jarboe, L.R.; Zhang, X.; Wang, X.; Moore, J.C.; Shanmugam, K.T.; Ingram, L.O. Metabolic engineering for production of biorenewable fuels and chemicals: Contributions of synthetic biology. J. Biomed. Biotechnol. 2010, 2010, 761042. [Google Scholar] [CrossRef]
  78. Da Silva, T.L.; Gouveia, L.; Reis, A. Integrated microbial processes for biofuels and high value-added products: The way to improve the cost effectiveness of biofuel production. Appl. Microbiol. Biotechnol. 2014, 98, 1043–1053. [Google Scholar] [CrossRef]
  79. Colin, V.L.; Rodríguez, A.; Cristóbal, H.A. The role of synthetic biology in the design of microbial cell factories for biofuel production. J. Biomed. Biotechnol. 2011, 2011, 601834. [Google Scholar] [CrossRef]
  80. Navale, G.R.; Dharne, M.S.; Shinde, S.S. Metabolic engineering and synthetic biology for isoprenoid production in Escherichia coli and Saccharomyces cerevisiae. Appl. Microbiol. Biotechnol. 2021, 105, 457–475. [Google Scholar] [CrossRef]
  81. Wang, J.; Li, C.; Jiang, T.; Yan, Y. Biosensor-assisted titratable CRISPRi high-throughput (BATCH) screening for over-production phenotypes. Metab. Eng. 2023, 75, 58–67. [Google Scholar] [CrossRef]
  82. Liu, W.; Peng, J.; Zou, S.; Xu, L.; Cheng, H.; Wang, Y.; Chen, Z.; Zhou, H. Regulation on pathway metabolic fluxes to enhance colanic acid production in Escherichia coli. J. Agric. Food Chem. 2023, 71, 13857–13868. [Google Scholar] [CrossRef] [PubMed]
  83. Wu, Z.; Chen, T.; Sun, W.; Chen, Y.; Ying, H. Optimizing Escherichia coli strains and fermentation processes for enhanced L-lysine production: A review. Front. Microbiol. 2024, 15, 1485624. [Google Scholar] [CrossRef]
  84. Koppolu, V.; Vasigala, V.K. Role of Escherichia coli in Biofuel Production. Microbiol. Insights 2016, 9, 29–35. [Google Scholar] [CrossRef]
  85. Wang, C.; Chang, D.; Zhang, Q.; Yu, Z. Enhanced bioethanol production by evolved Escherichia coli LGE2-H in a microbial electrolysis cell system. Bioresour. Bioprocess. 2024, 11, 4. [Google Scholar] [CrossRef] [PubMed]
  86. Hussain, M.H.; Sajid, S.; Martuscelli, M.; Aldahmash, W.; Mohsin, M.Z.; Ashraf, K.; Guo, M.; Mohsin, A. Sustainable biosynthesis of lycopene by using evolutionary adaptive recombinant Escherichia coli from orange peel waste. Heliyon 2024, 10, e34567. [Google Scholar] [CrossRef]
  87. Deepika, B.; Agnishwar, G.; Koyeli, G. Antioxidant and anticancer activity of nano lycopene. Res. J. Biotechnol. 2023, 18, 6. [Google Scholar]
  88. Zhao, L.; Zhu, Y.; Jia, H.; Han, Y.; Zheng, X.; Wang, M.; Feng, W. From plant to yeast—Advances in biosynthesis of artemisinin. Molecules 2022, 27, 6888. [Google Scholar] [CrossRef]
  89. Mittermeier, F.; Bäumler, M.; Arulrajah, P.; García Lima, J.J.; Hauke, S.; Stock, A.; Weuster-Botz, D. Artificial microbial consortia for bioproduction processes. Eng. Life Sci. 2023, 23, e2100152. [Google Scholar] [CrossRef]
  90. Li, X.; Wu, S.; Dong, Y.; Fan, H.; Bai, Z.; Zhuang, X. Engineering microbial consortia towards bioremediation. Water 2021, 13, 2928. [Google Scholar] [CrossRef]
  91. Meier, A.B.; Oppermann, S.; Drake, H.L.; Schmidt, O. Organic carbon from graminoid roots as a driver of fermentation in a fen. FEMS Microbiol. Ecol. 2021, 97, fiab143. [Google Scholar] [CrossRef] [PubMed]
  92. Li, D.; Meng, X.; Sun, Y.; Huang, R.; Liu, X.; Xie, Z.; Cao, Q. The microbial and functional reconstruction of instable syntrophic propionate-oxidizing methanogenesis by system recovering and injection modes changing. Chem. Eng. J. 2025, 455, 140736. [Google Scholar] [CrossRef]
  93. Wu, D.; Xie, W.; Li, X.; Cai, G.; Lu, J.; Xie, G. Metabolic engineering of Saccharomyces cerevisiae using the CRISPR/Cas9 system to minimize ethyl carbamate accumulation during Chinese rice wine fermentation. Appl. Microbiol. Biotechnol. 2020, 104, 4435–4444. [Google Scholar] [CrossRef]
  94. Chen, X.; Li, C.; Qiu, X.; Chen, M.; Xu, Y.; Li, S.; Li, Q.; Wang, L. CRISPR/Cas9-based iterative multi-copy integration for improved metabolite yields in Saccharomyces cerevisiae. Synth. Syst. Biotechnol. 2025, 10, 629–637. [Google Scholar] [CrossRef]
  95. Liu, H.; Zhou, P.; Qi, M.; Guo, L.; Gao, C.; Hu, G.; Song, W.; Wu, J.; Chen, X.; Chen, J. Enhancing biofuels production by engineering the actin cytoskeleton in Saccharomyces cerevisiae. Nat. Commun. 2022, 13, 1886. [Google Scholar] [CrossRef]
  96. Sun, L.; Jin, Y.S. Xylose assimilation for the efficient production of biofuels and chemicals by engineered Saccharomyces cerevisiae. Biotechnol. J. 2021, 16, 2000142. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, G.; Kell, D.B.; Borodina, I. Harnessing the yeast Saccharomyces cerevisiae for the production of fungal secondary metabolites. Essays Biochem. 2021, 65, 277–291. [Google Scholar] [PubMed]
  98. Bose, S.K.; Upadhyay, S.; Srivastava, Y. Metabolic Engineering of Microbes for the Production of Plant-Based Compounds. In Microbial Biotechnology: Role in Ecological Sustainability and Research; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2022; pp. 59–73. [Google Scholar]
  99. Liu, D. Improving Flavonoid Production in Saccharomyces cerevisiae Using Synthetic Biology Tools. Ph.D. Thesis, Chalmers Tekniska Hogskola, Gothenburg, Sweden, 2023. [Google Scholar]
  100. Patel, V.K.; Das, A.; Kumari, R.; Kajla, S. Recent progress and challenges in CRISPR-Cas9 engineered algae and cyanobacteria. Algal Res. 2023, 71, 103068. [Google Scholar] [CrossRef]
  101. Luan, G.; Zhang, S.; Lu, X. Engineering cyanobacteria chassis cells toward more efficient photosynthesis. Curr. Opin. Biotechnol. 2020, 62, 1–6. [Google Scholar] [CrossRef]
  102. Yao, L.; Shabestary, K.; Björk, S.M.; Asplund-Samuelsson, J.; Joensson, H.N.; Jahn, M.; Hudson, E.P. Pooled CRISPRi screening of the cyanobacterium Synechocystis sp. PCC 6803 for enhanced industrial phenotypes. Nat. Commun. 2020, 11, 1666. [Google Scholar] [CrossRef]
  103. Yadav, I.; Rautela, A.; Kumar, S. Approaches in the photosynthetic production of sustainable fuels by cyanobacteria using tools of synthetic biology. World J. Microbiol. Biotechnol. 2021, 37, 201. [Google Scholar] [CrossRef] [PubMed]
  104. Rodrigues, J.S.; Lindberg, P. Engineering cyanobacteria as host organisms for production of terpenes and terpenoids. In Cyanobacteria Biotechnology; Wiley: Hoboken, NJ, USA, 2021; pp. 267–300. [Google Scholar]
  105. Sitther, V.; Wyatt, L.; Jones, C.; Yalcin, Y. Bioactive compounds and pigments from cyanobacteria: Applications in the pharmaceutical industry. In Expanding Horizon of Cyanobacterial Biology; Academic Press: London, UK, 2022; pp. 65–90. [Google Scholar]
  106. Sarnaik, A.; Liu, A.; Nielsen, D.; Varman, A.M. High-throughput screening for efficient microbial biotechnology. Curr. Opin. Biotechnol. 2020, 64, 141–150. [Google Scholar] [CrossRef]
  107. O’Connor, E.; Micklefield, J.; Cai, Y. Searching for the optimal microbial factory: High-throughput biosensors and analytical techniques for screening small molecules. Curr. Opin. Biotechnol. 2024, 87, 103125. [Google Scholar] [CrossRef]
  108. Yeom, J.; Park, J.S.; Jung, S.-W.; Lee, S.; Kwon, H.; Yoo, S.M. High-throughput genetic engineering tools for regulating gene expression in a microbial cell factory. Crit. Rev. Biotechnol. 2023, 43, 82–99. [Google Scholar] [CrossRef]
  109. Madzak, C. Yarrowia lipolytica Strains and Their Biotechnological Applications: How Natural Biodiversity and Metabolic Engineering Could Contribute to Cell Factories Improvement. J. Fungi 2021, 7, 548. [Google Scholar] [CrossRef]
  110. Miranda, S.M.; Belo, I.; Lopes, M. Unraveling the Potential of Yarrowia lipolytica to Utilize Waste Motor Oil as a Carbon Source. J. Fungi 2024, 10, 777. [Google Scholar] [CrossRef] [PubMed]
  111. Lu, R.; Cao, L.; Wang, K.; Ledesma-Amaro, R.; Ji, X.-J. Engineering Yarrowia lipolytica to produce advanced biofuels: Current status and perspectives. Bioresour. Technol. 2021, 341, 125877. [Google Scholar] [CrossRef]
  112. Fabiszewska, A.; Paplińska-Goryca, M.; Misiukiewicz-Stępień, P.; Wołoszynowska, M.; Nowak, D.; Zieniuk, B. Expression Profile of Selected Genes Involved in Storage Lipid Synthesis in a Model Oleaginous Yeast Species Yarrowia lipolytica. Int. J. Mol. Sci. 2022, 23, 1041. [Google Scholar] [CrossRef]
  113. Qin, J.; Liu, N.; Abid, U.; Coleman, S.M.; Wang, Y.; Fu, Q.; Yoon, S.; Alper, H.S.; Xie, D. Metabolic Engineering of Yarrowia lipolytica for Conversion of Waste Cooking Oil into Omega-3 Eicosapentaenoic Acid. ACS Eng. Au 2025, 5, 128–139. [Google Scholar] [CrossRef]
  114. Ma, Y.; Liu, N.; Greisen, P.; Li, J.; Qiao, K.; Huang, S.; Stephanopoulos, G. Removal of lycopene substrate inhibition enables high carotenoid productivity in Yarrowia lipolytica. Nat. Commun. 2022, 13, 572. [Google Scholar] [CrossRef] [PubMed]
  115. Muhammad, A.; Feng, X.; Rasool, A.; Sun, W.; Li, C. Production of plant natural products through engineered Yarrowia lipolytica. Biotechnol. Adv. 2020, 43, 107555. [Google Scholar] [CrossRef]
  116. Zeng, B.X.; Yao, M.D.; Wang, Y.; Xiao, W.H.; Yuan, Y.J. Metabolic Engineering of Saccharomyces cerevisiae for Enhanced Dihydroartemisinic Acid Production. Front. Bioeng. Biotechnol. 2020, 8, 152. [Google Scholar] [CrossRef] [PubMed]
  117. Cunha, J.T.; Soares, P.O.; Baptista, S.L.; Costa, C.E.; Domingues, L. Engineered Saccharomyces cerevisiae for lignocellulosic valorization: A review and perspectives on bioethanol production. Bioengineered 2020, 11, 883–903. [Google Scholar] [CrossRef]
  118. Li, M.; Kildegaard, K.R.; Chen, Y.; Rodriguez, A.; Borodina, I.; Nielsen, J. De novo production of resveratrol from glucose or ethanol by engineered Saccharomyces cerevisiae. Metab. Eng. 2015, 32, 1–11. [Google Scholar] [CrossRef]
  119. Berla, B.M.; Saha, R.; Immethun, C.M.; Maranas, C.D.; Moon, T.S.; Pakrasi, H. Synthetic biology of cyanobacteria: Unique challenges and opportunities. Front. Microbiol. 2013, 4, 246. [Google Scholar] [CrossRef] [PubMed]
  120. Xie, D.; Jackson, E.N.; Zhu, Q. Sustainable source of omega-3 eicosapentaenoic acid from metabolically engineered Yarrowia lipolytica: From fundamental research to commercial production. Appl. Microbiol. 2015, 99, 1599–1610. [Google Scholar] [CrossRef]
  121. Nirmal, N.P.; Khanashyam, A.C.; Mundanat, A.S.; Shah, K.; Babu, K.S.; Thorakkattu, P.; Al-Asmari, F.; Pandiselvam, R. Valorization of Fruit Waste for Bioactive Compounds and Their Applications in the Food Industry. Foods 2023, 12, 556. [Google Scholar] [CrossRef]
  122. Gupta, P.; Sahoo, P.; Sandipam, S.; Gupta, R.P.; Kumar, M. Fermentation of biodiesel-derived crude glycerol to 1,3-propanediol with bio-wastes as support matrices: Polynomial prediction model. Enzyme Microb. Technol. 2023, 170, 110292. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, Y.; Liang, L.; Zheng, Y.; Han, J.; Xiu, Z. Improvement of 1,3-propanediol production from crude glycerol by co-cultivation of anaerobic and facultative microbes under non-strictly anaerobic conditions. Biotechnol. Biofuels Bioprod. 2022, 15, 40. [Google Scholar] [CrossRef]
  124. Mehariya, S.; Signorini, A.; Marone, A.; Rosa, S. Simultaneous hydrogen and ethanol production from crude glycerol by a microbial consortium using fed-batch fermentation. Energies 2023, 16, 4490. [Google Scholar] [CrossRef]
  125. Fan, W.; Huang, X.; Liu, K.; Xu, Y.; Hu, B.; Chi, Z. Nutrition component adjustment of distilled dried grain with Solubles via Aspergillus niger and its change about dynamic physiological metabolism. Fermentation 2022, 8, 264. [Google Scholar] [CrossRef]
  126. Yin, Z.; Liu, M.; Wang, B.; Zhao, D.; Li, H.; Sun, J. Extraction, identification, and in vitro anti-inflammatory activity of feruloylated oligosaccharides from Baijiu Distillers’ grains. Foods 2024, 13, 1283. [Google Scholar] [CrossRef] [PubMed]
  127. Troilo, M.; Difonzo, G.; Paradiso, V.M.; Summo, C.; Caponio, F. Bioactive compounds from vine shoots, grape stalks, and wine lees: Their potential use in agro-food chains. Foods 2021, 10, 342. [Google Scholar] [CrossRef]
  128. Athanasiou, P.E.; Patila, M.; Fotiadou, R.; Chatzikonstantinou, A.V.; Stamatis, H. Valorization of wine lees: Assessment of antioxidant, antimicrobial and enzyme inhibitory activity of wine lees extract and incorporation in chitosan films. Waste Biomass Valorization 2024, 15, 5657–5672. [Google Scholar] [CrossRef]
  129. Kriisa, M.; Taivosalo, A.; Föste, M.; Kütt, M.-L.; Viirma, M.; Priidik, R.; Korzeniowska, M.; Tian, Y.; Laaksonen, O.; Yang, B. Effect of enzyme-assisted hydrolysis on brewer’s spent grain protein solubilization–peptide composition and sensory properties. Appl. Food Res. 2022, 2, 100108. [Google Scholar] [CrossRef]
  130. Yang, L.; Li, X.; Lai, C.; Fan, Y.; Ouyang, J.; Yong, Q. Fungal chitosan production using xylose rich of corn stover prehydrolysate by Rhizopus oryzae. Biotechnol. Biotechnol. Equip. 2017, 31, 1160–1166. [Google Scholar] [CrossRef]
  131. Resmi, R.; Yoonus, J.; Beena, B. Anticancer and antibacterial activity of chitosan extracted from shrimp shell waste. Mater. Today Proc. 2021, 41, 570–576. [Google Scholar] [CrossRef]
  132. Sebastian, J.; Rouissi, T.; Brar, S.K.; Hegde, K.; Verma, M. Microwave-assisted extraction of chitosan from Rhizopus oryzae NRRL 1526 biomass. Carbohydr. Polym. 2019, 219, 431–440. [Google Scholar] [CrossRef]
  133. Lam, I.L.J.; Mohd Affandy, M.A.; ‘Aqilah, N.M.N.; Vonnie, J.M.; Felicia, W.X.L.; Rovina, K. Physicochemical characterization and antimicrobial analysis of vegetal chitosan extracted from distinct forest fungi species. Polymers 2023, 15, 2328. [Google Scholar] [CrossRef]
  134. Irbe, I.; Andze, L.; Blumfelde, M.; Filipova, I.; Verovkins, A.; Zoldners, J. Harvesting mycelial biomass of selected basidiomycetes for chitosan biopolymer extraction. Polymers 2023, 15, 3548. [Google Scholar] [CrossRef]
  135. Chen, L.-C.; Lin, S.-Y.; Sheu, M.-T.; Su, C.-H.; Lin, H.-L.; Hsieh, C.-M. Fabrication and characterization of Rhizochitosan and its incorporation with platelet concentrates to promote wound healing. Carbohydr. Polym. 2021, 268, 118239. [Google Scholar] [CrossRef] [PubMed]
  136. Pérez-Pérez, A.; Gullón, B.; Lobato-Rodríguez, Á.; Garrote, G.; Del Río, P.G. Microwave-assisted extraction of hemicellulosic oligosaccharides and phenolics from Robinia pseudoacacia wood. Carbohydr. Polym. 2023, 301, 120364. [Google Scholar] [CrossRef]
  137. Pereira, M.J.; Pedrosa, S.S.; Costa, J.R.; Carvalho, M.J.; Neto, T.; Oliveira, A.L.; Pintado, M.; Madureira, A.R. Sugarcane Straw Hemicellulose Extraction by Autohydrolysis for Cosmetic Applications. Molecules 2025, 30, 1208. [Google Scholar] [CrossRef]
  138. Gautério, G.V.; Amorim, C.; Silvério, S.C.; Cardoso, B.B.; Ballesteros, L.F.; Alves, J.I.; Pereira, M.A.; Silva, S.P.; Coelho, E.; Coimbra, M.A. Hydrolysates containing xylooligosaccharides produced by different strategies: Structural characterization, antioxidant and prebiotic activities. Food Chem. 2022, 391, 133231. [Google Scholar] [CrossRef]
  139. Li, F.; Li, Q.; Zhang, Y.; Zhou, X.; Yi, R.; Zhao, X. Effects of Xylooligosaccharides on Lipid Metabolism, Inflammation, and Gut Microbiota in C57BL/6J Mice Fed a High-Fat Diet. Front. Pharmacol. 2021, 12, 791614. [Google Scholar] [CrossRef] [PubMed]
  140. Cabeza Sánchez, Á.; Trygve Berglihn, O.; Ottaviano, E.; Rücker, T.; Pettersen, T.; Wittgens, B.; Aliko, A.; Gálvez, L.; López, M. Innovative vanillin yielding from lignin: Process modelling and assessment. Open Res. Eur. 2024, 4, 5. [Google Scholar] [CrossRef]
  141. García-Rollán, M.; Rivas-Márquez, M.N.; Bertran-Llorens, S.; Deuss, P.J.; Ruiz-Rosas, R.; Rosas, J.M.; Rodríguez-Mirasol, J.; Cordero, T. Biobased vanillin production by oxidative depolymerization of kraft lignin on a nitrogen-and phosphorus-functionalized activated carbon catalyst. Energy Fuels 2024, 38, 7018–7032. [Google Scholar] [CrossRef]
  142. Grinco, M.; Barba, A.; Kulciţki, V. Extraction of pharmaceutical grade lignins and their ozonolytic cleavage in a deep eutectic solvent. In New Frontiers in Natural Product Chemistry; Rahman, A., Ed.; Bentham Science Publishers: Singapore, 2022; p. 28. [Google Scholar]
  143. Karagoz, P.; Khiawjan, S.; Marques, M.P.C.; Santzouk, S.; Bugg, T.D.H.; Lye, G.J. Pharmaceutical applications of lignin-derived chemicals and lignin-based materials: Linking lignin source and processing with clinical indication. Biomass Convers. Biorefin. 2024, 14, 26553–26574. [Google Scholar] [CrossRef]
  144. de Souza Silva, A.P.; de Camargo, A.C.; Lazarini, J.G.; Franchin, M.; Sardi, J.C.O.; Rosalen, P.L.; de Alencar, S.M. Phenolic Profile and the Antioxidant, Anti-Inflammatory, and Antimicrobial Properties of Açaí (Euterpe oleracea) Meal: A Prospective Study. Foods 2022, 12, 86. [Google Scholar] [CrossRef]
  145. Salem, Y.; Sunoqrot, S.; Hammad, A.; Rajha, H.N.; Alzaghari, L.F.; Abusulieh, S.; Maroun, R.G.; Louka, N. Oxidation-Driven Assembly of Phenolic Compounds from Grape Seeds Waste into Nanoparticles as Potential Anti-Inflammatory and Wound Healing Therapies. ACS Appl. Bio Mater 2025, in press. [CrossRef] [PubMed]
  146. Panzella, L.; Moccia, F.; Nasti, R.; Marzorati, S.; Verotta, L.; Napolitano, A. Bioactive Phenolic Compounds From Agri-Food Wastes: An Update on Green and Sustainable Extraction Methodologies. Front. Nutr. 2020, 7, 60. [Google Scholar] [CrossRef]
  147. Nan, N.; Hu, W.; Wang, J. Lignin-based porous biomaterials for medical and pharmaceutical applications. Biomedicines 2022, 10, 747. [Google Scholar] [CrossRef] [PubMed]
  148. Katiyar, R.; Banerjee, S.; Arora, A. Recent advances in the integrated biorefinery concept for the valorization of algal biomass through sustainable routes. Biofuels Bioprod. Biorefin. 2021, 15, 879–898. [Google Scholar] [CrossRef]
  149. Premaratne, M.; Nishshanka, G.K.S.H.; Anthonio, R.A.D.P.; Liyanaarachchi, V.C.; Thevarajah, B.; Nimarshana, P.; Malik, A.; Ariyadasa, T.U. Resource recovery from waste streams for production of microalgae biomass: A sustainable approach towards high-value biorefineries. Bioresour. Technol. Rep. 2022, 18, 101070. [Google Scholar] [CrossRef]
  150. Kalasariya, H.S.; Pereira, L. Role of Seaweed as a Functional Ingredients in Nutraceuticals, Pharmaceuticals, Cosmetics, and Edible Salts. In Recent Advances in Seaweed Biotechnology: Biomass, Emerging Applications and Bioeconomy; Springer: Berlin/Heidelberg, Germany, 2025; pp. 347–390. [Google Scholar]
  151. Perez-Vazquez, A.; Carpena, M.; Barciela, P.; Cassani, L.; Simal-Gandara, J.; Prieto, M.A. Pressurized liquid extraction for the recovery of bioactive compounds from seaweeds for food industry application: A review. Antioxidants 2023, 12, 612. [Google Scholar] [CrossRef] [PubMed]
  152. Martins, A.; Silva, J.; Alves, C.; Pinteus, S.; Félix, C.; Augusto, A.; Pedrosa, R.; Mestre, A.S.; Santos, R.M.; Carvalho, A.P. Towards a zero-waste sustainable biorefinery of Codium sp. seaweed: From bioactives application to soil enhancement materials. J. Clean. Prod. 2024, 453, 142191. [Google Scholar] [CrossRef]
  153. Paul, V.; Agarwal, A.; Dutt Tripathi, A.; Sirohi, R. Valorization of lignin for the production of vanillin by Bacillus aryabhattai NCIM 5503. Bioresour. Technol. 2023, 385, 129420. [Google Scholar] [CrossRef]
  154. Valladares-Diestra, K.K.; de Souza Vandenberghe, L.P.; Vieira, S.; Goyzueta-Mamani, L.D.; de Mattos, P.B.G.; Manzoki, M.C.; Soccol, V.T.; Soccol, C.R. The Potential of Xylooligosaccharides as Prebiotics and Their Sustainable Production from Agro-Industrial by-Products. Foods 2023, 12, 2681. [Google Scholar] [CrossRef]
  155. Bai, R.; Nguyen, T.T.; Zhou, Y.; Diao, Y.; Zhang, W. Identification of Antioxidative Peptides Derived from Arthrospira maxima in the Biorefinery Process after Extraction of C-Phycocyanin and Lipids. Mar. Drugs 2023, 21, 146. [Google Scholar] [CrossRef]
  156. Santana, Á.L.; Meireles, M.A.A. Valorization of Cereal Byproducts with Supercritical Technology: The Case of Corn. Fermentation 2023, 11, 289. [Google Scholar] [CrossRef]
  157. Baharlouei, P.; Rahman, A. Chitin and Chitosan: Prospective Biomedical Applications in Drug Delivery, Cancer Treatment, and Wound Healing. Mar. Drugs 2022, 20, 460. [Google Scholar] [CrossRef] [PubMed]
  158. Pramanik, A.; Chaudhary, A.A.; Sinha, A.; Chaubey, K.K.; Ashraf, M.S.; Basher, N.S.; Rudayni, H.A.; Dayal, D.; Kumar, S. Nanocatalyst-Based Biofuel Generation: An Update, Challenges and Future Possibilities. Sustainability 2023, 15, 6180. [Google Scholar] [CrossRef]
  159. Akia, M.; Yazdani, F.; Motaee, E.; Han, D.; Arandiyan, H. A review on conversion of biomass to biofuel by nanocatalysts. Biofuel Res. J. 2014, 1, 16–25. [Google Scholar] [CrossRef]
  160. Sun, Y.; Ma, Y.; Zhang, B.; Sun, H.; Wang, N.; Wang, L.; Zhang, J.; Xue, R. Comparison of magnetite/reduced graphene oxide nanocomposites and magnetite nanoparticles on enhancing hydrogen production in dark fermentation. Int. J. Hydrogen Energy 2022, 47, 22359–22370. [Google Scholar] [CrossRef]
  161. Ingle, A.P.; Chandel, A.K.; Philippini, R.; Martiniano, S.E.; da Silva, S.S. Advances in nanocatalysts mediated biodiesel production: A critical appraisal. Symmetry 2020, 12, 256. [Google Scholar] [CrossRef]
  162. Montiel Schneider, M.G.; Martín, M.J.; Otarola, J.; Vakarelska, E.; Simeonov, V.; Lassalle, V.; Nedyalkova, M. Biomedical applications of iron oxide nanoparticles: Current insights progress and perspectives. Pharmaceutics 2022, 14, 204. [Google Scholar] [CrossRef]
  163. Dos Santos, K.M.; de França Serpa, J.; de Castro Bizerra, V.; Melo, R.L.F.; Sousa Junior, P.G.a.d.; Santos Alexandre, V.; da Fonseca, A.M.; Fechine, P.B.A.; Lomonaco, D.; Sousa dos Santos, J.C. Enhanced Biodiesel Production with Eversa Transform 2.0 Lipase on Magnetic Nanoparticles. Langmuir 2024, 40, 26835–26851. [Google Scholar] [CrossRef]
  164. Krishnan, P.D.; Jayavignesh, V.; Ramkumar, G.; Purushothaman, Y.; Sadhasivam, S.; Sabarathinam, S.; Venkatesan, K.; Sivakumar, E.; Ganesh, R.S.; Swaminathan, K. Synthesis, surface modification, and application of iron oxide nanoparticles in lipase immobilization for biodiesel production: A review. In Nanomaterials; Apple Academic Press: New York, NY, USA, 2018. [Google Scholar]
  165. Karnwal, A.; Kumar Sachan, R.S.; Devgon, I.; Devgon, J.; Pant, G.; Panchpuri, M.; Ahmad, A.; Alshammari, M.B.; Hossain, K.; Kumar, G. Gold nanoparticles in nanobiotechnology: From synthesis to biosensing applications. ACS Omega 2024, 9, 29966–29982. [Google Scholar] [CrossRef] [PubMed]
  166. Hu, X.; Zhang, Y.; Ding, T.; Liu, J.; Zhao, H. Multifunctional gold nanoparticles: A novel nanomaterial for various medical applications and biological activities. Front. Bioeng. Biotechnol. 2020, 8, 990. [Google Scholar] [CrossRef]
  167. Gupta, K.; Rai, R.K.; Singh, S.K. Metal catalysts for the efficient transformation of biomass-derived HMF and furfural to value added chemicals. ChemCatChem 2018, 10, 2326–2349. [Google Scholar] [CrossRef]
  168. Wang, D.; Li, G.; Zhang, C.; Wang, Z.; Li, X. Nickel nanoparticles inlaid in lignin-derived carbon as high effective catalyst for lignin depolymerization. Bioresour. Technol. 2019, 289, 121629. [Google Scholar] [CrossRef] [PubMed]
  169. Awogbemi, O.; Ojo, A.A.; Adeleye, S.A. Advancements in the application of metal oxide nanocatalysts for sustainable biodiesel production. Discov. Appl. Sci. 2024, 6, 250. [Google Scholar] [CrossRef]
  170. Yassin, M.T.; Al-Otibi, F.O.; Al-Sahli, S.A.; El-Wetidy, M.S.; Mohamed, S. Metal Oxide Nanoparticles as Efficient Nanocarriers for Targeted Cancer Therapy: Addressing Chemotherapy-Induced Disabilities. Cancers 2024, 16, 4234. [Google Scholar] [CrossRef]
  171. Mandal, A.K.; Katuwal, S.; Tettey, F.; Gupta, A.; Bhattarai, S.; Jaisi, S.; Bhandari, D.P.; Shah, A.K.; Bhattarai, N.; Parajuli, N. Current research on zinc oxide nanoparticles: Synthesis, characterization, and biomedical applications. Nanomaterials 2022, 12, 3066. [Google Scholar] [CrossRef]
  172. Ashour, M.; Mansour, A.T.; Abdelwahab, A.M.; Alprol, A.E. Metal oxide nanoparticles’ green synthesis by plants: Prospects in phyto-and bioremediation and photocatalytic degradation of organic pollutants. Processes 2023, 11, 3356. [Google Scholar] [CrossRef]
  173. Sharma, R.K.; Bandichhor, R.; Mishra, V.; Sharma, S.; Yadav, S.; Mehta, S.; Arora, B.; Rana, P.; Dutta, S.; Solanki, K. Advanced metal oxide-based nanocatalysts for the oxidative synthesis of fine chemicals. Mater. Adv. 2023, 4, 1795–1830. [Google Scholar] [CrossRef]
  174. Li, Y.; Zhang, Y.; Qian, K.; Huang, W. Metal–support interactions in metal/oxide catalysts and oxide–metal interactions in oxide/metal inverse catalysts. ACS Catal. 2022, 12, 1268–1287. [Google Scholar] [CrossRef]
  175. AbdulRasheed, T.; Afotey, B.; Ankudey, E.G.; Anang, D.A. Synthesis and characterization of novel calcium oxide/calcium ferrite, CaO/CaFe2O4 composite nanocatalyst for biodiesel production. ES Mater. Manuf. 2023, 22, 922. [Google Scholar] [CrossRef]
  176. Alonso, D.M.; Wettstein, S.G.; Dumesic, J.A. Bimetallic catalysts for upgrading of biomass to fuels and chemicals. Chem. Soc. Rev. 2012, 41, 8075–8098. [Google Scholar] [CrossRef]
  177. Pelicano, C.M.; Saruyama, M.; Takahata, R.; Sato, R.; Kitahama, Y.; Matsuzaki, H.; Yamada, T.; Hisatomi, T.; Domen, K.; Teranishi, T. Bimetallic synergy in ultrafine cocatalyst alloy nanoparticles for efficient photocatalytic water splitting. Adv. Funct. Mater. 2022, 32, 2202987. [Google Scholar] [CrossRef]
  178. Liu, Q.; Han, X.; Zheng, Z.; Xiong, P.; Jeong, R.G.; Kim, G.; Park, H.; Kim, J.; Kim, B.K.; Park, H.S. Crystallinity regulated functional separator based on bimetallic NixFey alloy nanoparticles for facilitated redox kinetics of lithium–sulfur batteries. Adv. Funct. Mater. 2022, 32, 2207094. [Google Scholar] [CrossRef]
  179. Mustieles Marin, I.; Asensio, J.M.; Chaudret, B. Bimetallic nanoparticles associating noble metals and first-row transition metals in catalysis. ACS Nano 2021, 15, 3550–3556. [Google Scholar] [CrossRef]
  180. Sharif, A.; Khan, S.M.; Gull, N.; Rizwan, K.; Munir, S.; Shakeel, M.; Islam, A. Introduction to nanomaterials: A strategic tool for production of biofuel and bioenergy from biomass. In Nanomaterials in Biomass Conversion; Elsevier: Amsterdam, The Netherlands, 2024; pp. 57–84. [Google Scholar]
  181. Guo, G.; Li, W.; Dou, X.; Ogunbiyi, A.T.; Ahmed, T.; Zhang, B.; Wu, M. Hydroconversion of Kraft lignin for biofuels production using bifunctional rhenium-molybdenum supported zeolitic imidazolate framework nanocatalyst. Bioresour. Technol. 2021, 321, 124443. [Google Scholar] [CrossRef]
  182. Usman, M.A.; Naeem, M.; Saeed, M.; Zaheer, M. Catalytic C–O bond cleavage in a β-O-4 lignin model through intermolecular hydrogen transfer. Inorg. Chim. Acta 2021, 521, 120305. [Google Scholar] [CrossRef]
  183. Wang, Y.; Wei, L.; Hou, Q.; Mo, Z.; Liu, X.; Li, W. A review on catalytic depolymerization of lignin towards high-value chemicals: Solvent and catalyst. Fermentation 2023, 9, 386. [Google Scholar] [CrossRef]
  184. Blay-Roger, R.; Carrasco-Ruiz, S.; Reina, T.R.; Bobadilla, L.F.; Odriozola, J.A.; Nawaz, M.A. Integrating catalytic tandem reactions for the next generation of biofuels: A perspective. Chem. Catal. 2024, 4, 100897. [Google Scholar] [CrossRef]
  185. Merkouri, L.-P.; Paksoy, A.I.; Ramirez Reina, T.; Duyar, M.S. The need for flexible chemical synthesis and how dual-function materials can pave the way. ACS Catal. 2023, 13, 7230–7242. [Google Scholar] [CrossRef]
  186. Mullins, L.; Sullivan, J.A. Synthesis of a Sustainable Cellulose-Derived Biofuel Through a 1-Pot, 2-Catalyst Tandem Reaction. Top. Catal. 2020, 63, 1434–1445. [Google Scholar] [CrossRef]
  187. Song, Y.; Sun, P.; Qiu, W.; Li, Y.; Peng, C. Catalytic and engineering strategies for enhanced hydrogenation reactions: A review of heterogeneous catalysts and process optimization. Results Eng. 2025, 25, 103958. [Google Scholar] [CrossRef]
  188. Akram, H.A.; Imran, M.; Javaid, A.; Latif, S.; Rizvi, N.B.; Jesionowski, T.; Bilal, M. Pretreatment and catalytic conversion of lignocellulosic and algal biomass into biofuels by metal organic frameworks. Mol. Catal. 2023, 539, 112893. [Google Scholar] [CrossRef]
  189. Zhou, Z.W.; Cai, C.X.; Xing, X.; Li, J.; Hu, Z.E.; Xie, Z.B.; Wang, N.; Yu, X.Q. Magnetic COFs as satisfied support for lipase immobilization and recovery to effectively achieve the production of biodiesel by great maintenance of enzyme activity. Biofuels 2021, 14, 156. [Google Scholar]
  190. Silva, F.M.W.G.; Szemes, J.; Mustashev, A.; Takács, O.; Imarah, A.O.; Poppe, L. Immobilization of Lipase B from Candida antarctica on Magnetic Nanoparticles Enhances Its Selectivity in Kinetic Resolutions of Chiral Amines with Several Acylating Agents. Fermentation 2023, 13, 1560. [Google Scholar] [CrossRef]
  191. Duan, Y.; Cheng, Y.; Hu, Z.; Wang, C.; Sui, D.; Yang, Y.; Lu, T. A Comprehensive Review on Metal Catalysts for the Production of Cyclopentanone Derivatives from Furfural and HMF. Molecules 2023, 28, 5397. [Google Scholar] [CrossRef]
  192. Ronel, D.; Nasya, N.; Duarte, J.; Cheng, Y.-S.; Selvasembian, R.; Amir, F.; De Oliveira, L.; Wan Azelee, N.I.; Meili, L.; Rangasamy, G. A comprehensive review on nanocatalysts and nanobiocatalysts for biodiesel production in Indonesia, Malaysia, Brazil and USA. Chemosphere 2023, 319, 138003. [Google Scholar]
  193. Jeevanandam, J.; Manchala, S.; Danquah, M.K. Wastewater Treatment by Photocatalytic Biosynthesized Nanoparticles. In Handbook of Nanomaterials and Nanocomposites for Energy and Environmental Applications, 1st ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 1–23. [Google Scholar]
  194. Bronstein, L.M.; Matveeva, V.G. Multifunctional Catalysts for Cascade Reactions in Biomass Processing. Fermentation 2024, 14, 1937. [Google Scholar] [CrossRef] [PubMed]
  195. Sanz, A.; Susmozas, A.; Peters, J.; Dufour, J. Biorefinery Modeling and Optimization. In Biorefineries; Rabaçal, M., Ferreira, A., Silva, C., Costa, M., Eds.; Springer: Cham, Switzerland, 2017; Volume 57, pp. 123–160. [Google Scholar]
  196. Chemmangattuvalappil, N.G.; Ng, D.K.S.; Ng, L.Y.; Ooi, J.; Chong, J.W.; Eden, M.R. A Review of Process Systems Engineering (PSE) Tools for the Design of Ionic Liquids and Integrated Biorefineries. Processes 2020, 8, 1678. [Google Scholar] [CrossRef]
  197. Shen, F.; Sun, R.; Yao, J.; Li, J.; Liu, Q.; Price, N.D.; Liu, C.; Wang, Z. OptRAM: In-silico strain design via integrative regulatory-metabolic network modeling. PLoS Comput. Biol. 2019, 15, e1006835. [Google Scholar] [CrossRef] [PubMed]
  198. Raman, K.; Chandra, N. Flux balance analysis of biological systems: Applications and challenges. Brief. Bioinform. 2009, 10, 435–449. [Google Scholar] [CrossRef]
  199. Raajaraam, L.; Raman, K. A Computational Framework to Identify Metabolic Engineering Strategies for the Co-Production of Metabolites. Front. Bioeng. Biotechnol. 2022, 9, 779405. [Google Scholar] [CrossRef]
  200. Carbonell, P.; Parutto, P.; Baudier, C.; Junot, C.; Faulon, J.-L. Retropath: Automated Pipeline for Embedded Metabolic Circuits. ACS Synth. Biol. 2014, 3, 565–577. [Google Scholar] [CrossRef]
  201. Moura, M.; Broadbelt, L.; Tyo, K. Computational Tools for Guided Discovery and Engineering of Metabolic Pathways. In Systems Metabolic Engineering: Methods and Protocols; Alper, H.S., Ed.; Humana Press: Totowa, NJ, USA, 2013; Volume 985, pp. 123–147. [Google Scholar]
  202. Lopes, T.F.; Ortigueira, J.; Matos, C.T.; Costa, L.; Ribeiro, C.; Reis, A.; Gírio, F. Conceptual Design of an Autotrophic Multi-Strain Microalgae-Based Biorefinery: Preliminary Techno-Economic and Life Cycle Assessments. Fermentation 2023, 9, 255. [Google Scholar] [CrossRef]
  203. Lawson, C.E.; Martí, J.M.; Radivojevic, T.; Jonnalagadda, S.V.R.; Gentz, R.; Hillson, N.J.; Peisert, S.; Kim, J.; Simmons, B.A.; Petzold, C.J.; et al. Machine learning for metabolic engineering: A review. Metab. Eng. 2021, 63, 34–60. [Google Scholar] [CrossRef]
  204. Lee, M.K.; Mohamad, M.S.; Choon, Y.W.; Mohd Daud, K.; Nasarudin, N.A.; Ismail, M.A.; Ibrahim, Z.; Napis, S.; Sinnott, R.O. Comparison of optimization-modelling methods for metabolites production in Escherichia coli. J. Integr. Bioinform. 2020, 17, 20190073. [Google Scholar] [CrossRef]
  205. Patané, A.; Jansen, G.; Conca, P.; Carapezza, G.; Costanza, J.; Nicosia, G. Multi-objective optimization of genome-scale metabolic models: The case of ethanol production. Ann. Oper. Res. 2019, 276, 211–227. [Google Scholar] [CrossRef]
  206. Mienda, B.S.; Dräger, A. Genome-scale metabolic modeling of Escherichia coli and its chassis design for synthetic biology applications. In Computational Methods in Synthetic Biology; Marchisio, M.A., Ed.; Springer: New York, NY, USA, 2021; pp. 217–229. [Google Scholar]
  207. Bernstein, D.B.; Sulheim, S.; Almaas, E.; Segrè, D. Addressing uncertainty in genome-scale metabolic model reconstruction and analysis. Genome Biol. 2021, 22, 64. [Google Scholar] [CrossRef] [PubMed]
  208. Qian, J.; Ye, C. Development and applications of genome-scale metabolic network models. Adv. Appl. Microbiol. 2024, 126, 1–26. [Google Scholar]
  209. Kulyashov, M.A.; Kolmykov, S.K.; Khlebodarova, T.M.; Akberdin, I.R. State-of the-art constraint-based modeling of microbial metabolism: From basics to context-specific models with a focus on methanotrophs. Microorganisms 2023, 11, 2987. [Google Scholar] [CrossRef]
  210. Knoop, H.; Steuer, R. A computational analysis of stoichiometric constraints and trade-offs in cyanobacterial biofuel production. Front. Bioeng. Biotechnol. 2015, 3, 47. [Google Scholar] [CrossRef] [PubMed]
  211. Antoniewicz, M.R. A guide to metabolic flux analysis in metabolic engineering: Methods, tools and applications. Metab. Eng. 2021, 63, 2–12. [Google Scholar] [CrossRef]
  212. Sen, P. Flux balance analysis of metabolic networks for efficient engineering of microbial cell factories. Biotechnol. Genet. Eng. Rev. 2024, 40, 3682–3715. [Google Scholar] [CrossRef]
  213. Griesemer, M.; Navid, A. Uses of multi-objective flux analysis for optimization of microbial production of secondary metabolites. Microorganisms 2023, 11, 2149. [Google Scholar] [CrossRef]
  214. Sundqvist, N.; Grankvist, N.; Watrous, J.; Mohit, J.; Nilsson, R.; Cedersund, G. Validation-based model selection for 13C metabolic flux analysis with uncertain measurement errors. PLoS Comput. Biol. 2022, 18, e1009999. [Google Scholar] [CrossRef] [PubMed]
  215. Sake, C.L.; Metcalf, A.J.; Meagher, M.; Di Paola, J.; Neeves, K.B.; Boyle, N.R. Isotopically nonstationary 13C metabolic flux analysis in resting and activated human platelets. Metab. Eng. 2022, 69, 313–322. [Google Scholar] [CrossRef]
  216. Kong, Y.; Chen, H.; Huang, X.; Chang, L.; Yang, B.; Chen, W. Precise metabolic modeling in post-omics era: Accomplishments and perspectives. Crit. Rev. Biotechnol. 2025, 45, 683–701. [Google Scholar] [CrossRef]
  217. Hafner, J.; Mohammadi-Peyhani, H.; Hatzimanikatis, V. Pathway Design. In Metabolic Engineering: Concepts and Applications, 1st ed.; Lee, S.Y., Nielsen, J., Stephanopoulos, G., Eds.; Wiley: Weinheim, Germany, 2021; Volume 13, pp. 237–257. [Google Scholar]
  218. Zheng, S.; Zeng, T.; Li, C.; Chen, B.; Coley, C.W.; Yang, Y.; Wu, R. Deep learning driven biosynthetic pathways navigation for natural products with BioNavi-NP. Nat. Commun. 2022, 13, 3342. [Google Scholar] [CrossRef]
  219. Sveshnikova, A.; MohammadiPeyhani, H.; Hatzimanikatis, V. Computational tools and resources for designing new pathways to small molecules. Curr. Opin. Biotechnol. 2022, 76, 102722. [Google Scholar] [CrossRef] [PubMed]
  220. Bhattacharjee, N.; Alonso-Cotchico, L.; Lucas, M.F. Enzyme immobilization studied through molecular dynamic simulations. Front. Bioeng. Biotechnol. 2023, 11, 1200293. [Google Scholar] [CrossRef] [PubMed]
  221. Childers, M.C.; Daggett, V. Insights from molecular dynamics simulations for computational protein design. Mol. Syst. Des. Eng. 2017, 2, 9–33. [Google Scholar] [CrossRef]
  222. Perilla, J.R.; Goh, B.C.; Cassidy, C.K.; Liu, B.; Bernardi, R.C.; Rudack, T.; Yu, H.; Wu, Z.; Schulten, K. Molecular dynamics simulations of large macromolecular complexes. Curr. Opin. Struct. Biol. 2015, 31, 64–74. [Google Scholar] [CrossRef]
  223. Hu, X.; Zeng, Z.; Zhang, J.; Wu, D.; Li, H.; Geng, F. Molecular dynamics simulation of the interaction of food proteins with small molecules. Food Chem. 2023, 405, 134824. [Google Scholar] [CrossRef]
  224. Rithuan, R.H.B. Simulation Study of Process Flow Design for Downstream Processing of Succinic Acid Production Using SuperPro Designer Software; Universiti Teknologi Malaysia: Johor Bahru, Malaysia, 2021. [Google Scholar]
  225. Esmaeili-Faraj, S.H.; Rezazadeh, M.; Abdi, J. Simulation of Biosynthesis Gas Process from Palm Oil Mill Effluent Sewage by Aspen HYSYS and SuperPro Designer. J. Water Wastewater 2023, 33, 34–43. [Google Scholar]
  226. Gómez, J.A.; Sánchez, Ó.J.; Correa, L.F. Techno-economic and environmental evaluation of cheesemaking waste valorization through process simulation using SuperPro designer. Waste Biomass Valorization 2020, 11, 6025–6045. [Google Scholar] [CrossRef]
  227. Jang, W.D.; Kim, G.B.; Kim, Y.; Lee, S.Y. Applications of artificial intelligence to enzyme and pathway design for metabolic engineering. Curr. Opin. Biotechnol. 2022, 73, 101–107. [Google Scholar] [CrossRef]
  228. Diment, D.; Löfgren, J.; Alopaeus, M.; Stosiek, M.; Cho, M.; Xu, C.; Hummel, M.; Rigo, D.; Rinke, P.; Balakshin, M. AI-guided biorefinery optimization for the production of lignin-carbohydrate complexes with tailored properties. ChemRxiv 2024, submitted.
  229. Arjmand, B.; Hamidpour, S.K.; Tayanloo-Beik, A.; Goodarzi, P.; Aghayan, H.R.; Adibi, H.; Larijani, B. Machine learning: A new prospect in multi-omics data analysis of cancer. Front. Genet. 2022, 13, 824451. [Google Scholar] [CrossRef] [PubMed]
  230. Khaleghi, M.K.; Savizi, I.S.P.; Lewis, N.E.; Shojaosadati, S.A. Synergisms of machine learning and constraint-based modeling of metabolism for analysis and optimization of fermentation parameters. Biotechnol. J. 2021, 16, 2100212. [Google Scholar] [CrossRef]
  231. Palacios, S.; Collins, J.J.; Del Vecchio, D. Machine learning for synthetic gene circuit engineering. Curr. Opin. Biotechnol. 2025, 92, 103263. [Google Scholar] [CrossRef] [PubMed]
  232. Zheng, S.; Shao, M.; Wang, W.; Chen, G.-Q. Next-generation biotechnology inspired by extremes. EMBO Rep. 2025, 26, 1191–1195. [Google Scholar] [CrossRef]
  233. Mohanta, S.; Bahuguna, M.; Baley, J.D.; Sharma, S.; Sharma, V. Extremophilic Cellulases: A Comprehensive Review. J. Trop. Biol. 2023, 8, 74986. [Google Scholar] [CrossRef]
  234. Dumorné, K.; Córdova, D.C.; Astorga-Eló, M.; Renganathan, P. Extremozymes: A Potential Source for Industrial Applications. J. Microbiol. Biotechnol. 2017, 27, 649–659. [Google Scholar] [CrossRef]
  235. Barbosa, M.; Garcia, I.L.; Clélia, A.; Maranhão, P. The microalga Dunaliella and its applications: A review. Appl. Phycol. 2023, 4, 99–120. [Google Scholar] [CrossRef]
  236. Giani, M.; Garbayo, I.; Vílchez, C.; Martínez-Espinosa, R.M. Haloarchaeal Carotenoids: Healthy Novel Compounds from Extreme Environments. Mar. Drugs 2019, 17, 524. [Google Scholar] [CrossRef]
  237. Priya, J.P.; Rautela, J.; Pandey, P.; Morris, S.; Ghildiyal, P. Salt Tolerant Microbes (Active Metabolites) Mediated Nanoparticle: Drug Delivery and Future Prospects. J. Pharm. Appl. Med. 2024, 18, 853–866. [Google Scholar] [CrossRef]
  238. Koller, M.; Rittmann, S. Haloarchaea as Emerging Big Players in Future Polyhydroxyalkanoate Bioproduction: Review of Trends and Perspectives. Curr. Res. Biotechnol. 2022, 4, 100–112. [Google Scholar] [CrossRef]
  239. Hamid, B.; Bashir, Z.; Yatoo, A.M.; Mohiddin, F.; Majeed, N.; Bansal, M.; Poczai, P.; Almalki, W.H.; Sayyed, R.Z.; Shati, A.A.; et al. Cold-Active Enzymes and Their Potential Industrial Applications—A Review. Molecules 2022, 27, 5885. [Google Scholar] [CrossRef] [PubMed]
  240. Nichols, D.S. Prokaryotes and the input of polyunsaturated fatty acids to the marine food web. FEMS Microbiol. Lett. 2003, 219, 1–7. [Google Scholar] [CrossRef] [PubMed]
  241. Mukhtar, S.; Aslam, M. Biofuel synthesis by extremophilic microorganisms. In Biofuels Production–Sustainability and Advances in Microbial Bioresources; Yadav, A.N., Rastegari, A.A., Yadav, N., Gaur, R., Eds.; Biofuel and Biorefinery Technologies; Springer: Cham, Switzerland, 2020; Volume 11, pp. 115–138. [Google Scholar]
  242. Ghosh, A. Hydrolases from Extremophiles: Exploring Alternative Strategies for Low-Cost Biofuel Production. In Biofuels; CRC Press: Boca Raton, FL, USA, 2024; pp. 440–463. [Google Scholar]
  243. Arbab, S.; Ullah, H.; Khan, M.I.; Khattak, M.N.; Zhang, J.; Li, K.; Hassan, I.U. Diversity and distribution of thermophilic microorganisms and their applications in biotechnology. J. Basic Microbiol. 2022, 62, 95–108. [Google Scholar] [CrossRef]
  244. Ajeje, S.B.; Hu, Y.; Song, G.; Peter, S.B.; Afful, R.G.; Sun, F.; Asadollahi, M.A.; Amiri, H.; Abdulkhani, A.; Sun, H. Thermostable cellulases/xylanases from thermophilic and hyperthermophilic microorganisms: Current perspective. Front. Bioeng. Biotechnol. 2021, 9, 794304. [Google Scholar] [CrossRef]
  245. Hashemi, A.; Pajoum Shariati, F.; Delavari Amrei, H.; Heydari Nasab, A. The effect of instantaneous and slow-release salt stress methods on beta-carotene production within Dunaliella salina Cells. Iran. J. Chem. Chem. Eng. 2021, 40, 1642–1652. [Google Scholar]
  246. Tuyen, V.T.X.; Khai, T.; Diem, N.T.T.; Xuan, L.N.T.; Tan, N.D. Effect of supplied salt concentrations in the nutrient solution during hydroponic production on phytochemicals and antioxidant activity of ice plants (Mesembryanthemum crystallinum L.). Int. J. Agric. Biosci. 2025, 14, 59–67. [Google Scholar]
  247. De Souza Celente, G.; de Cassia de Souza Schneider, R.; Julich, J.; Rizzetti, T.M.; Lobo, E.A.; Sui, Y. Life cycle assessment of microalgal cultivation medium: Biomass, glycerol, and beta-carotene production by Dunaliella salina and Dunaliella tertiolecta. Int. J. Life Cycle Assess. 2023, 29, 2269–2282. [Google Scholar] [CrossRef]
  248. Mohanta, A.; Khadim, S.R.; Singh, P.; Laxmi; Asthana, R. A Study on Mixotrophic Approach for Maximizing Lipid Production in a Hypersaline Microalga, Dunaliella salina. BioEnergy Res. 2023, 16, 2512–2528. [Google Scholar] [CrossRef]
  249. Sharma, Y.; Singh, G.P.; Singh, C.P. Biorefinery potential of Dunaliella (Chlorophyta) for production of value-added compounds. Biofuels Bioprod. Biorefin. 2024, 18, 1755–1781. [Google Scholar] [CrossRef]
  250. Nguyen, P.T.H.; Nguyen, D.T.; Vo, T. Determination of Carotenoid and Beta-Carotene Content in Dunaliella Microalgae Powder. Eur. J. Med. Health Res. 2025, 3, 21–26. [Google Scholar] [CrossRef]
  251. Moopantakath, J.; Imchen, M.; Kumavath, R.; Martínez-Espinosa, R.M. Ubiquitousness of Haloferax and carotenoid producing genes in Arabian sea coastal biosystems of India. Mar. drugs 2021, 19, 442. [Google Scholar] [CrossRef] [PubMed]
  252. Bouhamed, S.B.H.; Chaari, M.; Baati, H.; Zouari, S.; Ammar, E. Extreme halophilic Archaea: Halobacterium salinarum carotenoids characterization and antioxidant properties. Heliyon 2024, 10, e25843. [Google Scholar] [CrossRef] [PubMed]
  253. Favreau, C.; Tribondeau, A.; Marugan, M.; Guyot, F.; Alpha-Bazin, B.; Marie, A.; Puppo, R.; Dufour, T.; Huguet, A.; Zirah, S. Molecular acclimation of Halobacterium salinarum to halite brine inclusions. Front. Microbiol. 2023, 13, 1075274. [Google Scholar] [CrossRef] [PubMed]
  254. Cho, E.-S.; Cha, I.-T.; Roh, S.W.; Seo, M.-J. Haloferax litoreum sp. nov., Haloferax marinisediminis sp. nov., and Haloferax marinum sp. nov., low salt-tolerant haloarchaea isolated from seawater and sediment. Antonie Leeuwenhoek 2021, 114, 2065–2082. [Google Scholar] [CrossRef]
  255. Pfeifer, F. Recent advances in the study of gas vesicle proteins and application of gas vesicles in biomedical research. Life 2022, 12, 1455. [Google Scholar] [CrossRef]
  256. Kasirajan, L.; Maupin-Furlow, J.A. Halophilic archaea and their potential to generate renewable fuels and chemicals. Biotechnol. Bioeng. 2021, 118, 1066–1090. [Google Scholar] [CrossRef]
  257. Martínez-Espinosa, R.M. Halocins and C50 Carotenoids from Haloarchaea: Potential Natural Tools against Cancer. Mar. Drugs 2024, 22, 448. [Google Scholar] [CrossRef]
  258. Kumari, S.; Satapathy, S.; Datta, M.; Kumar, S. Adaptation of microalgae to temperature and light stress. In Plant Stress: Challenges and Management in the New Decade; Roy, S., Mathur, P., Chakraborty, A.P., Saha, S.P., Eds.; Advances in Science, Technology & Innovation; Springer: Cham, Switzerland, 2022; pp. 123–134. [Google Scholar]
  259. Kashyap, A.K.; Dubey, S.K.; Jain, B.P. Cyanobacterial diversity concerning the extreme environment and their bioprospecting. In Cyanobacterial Lifestyle and Its Applications in Biotechnology; Elsevier: Amsterdam, The Netherlands, 2022; pp. 1–22. [Google Scholar]
  260. Fernández-Sanromán, Á.; Sanromán, M.Á. Extremozymes in food production and processing. In Value-Addition in Food Products and Processing Through Enzyme Technology; Elsevier: Amsterdam, The Netherlands, 2022; pp. 25–43. [Google Scholar]
  261. Khan, N.F.; Zehra, U.; Reshi, Z.A.; Shah, M.A.; Baba, T.R. Seasonal Dynamics of Bacterial and Fungal Lineages in Extreme Environments. In Climate Change and Microbial Diversity; Apple Academic Press: New York, NY, USA, 2022; pp. 63–107. [Google Scholar]
  262. Quatrini, R.; Johnson, D.B. Acidithiobacillus ferrooxidans. Trends Microbiol. 2019, 27, 282–283. [Google Scholar] [CrossRef]
  263. Tonietti, L.; Esposito, M.; Cascone, M.; Barosa, B.; Fiscale, S.; Tomajoli, M.T.M.; Sbaffi, T.; Santomartino, R.; Covone, G.; Cordone, A. Unveiling the bioleaching versatility of Acidithiobacillus ferrooxidans. Microorganisms 2024, 12, 2407. [Google Scholar] [CrossRef]
  264. Wu, P.; Zhang, L.-J.; Lin, C.-B.; Xie, X.-X.; Yong, X.-Y.; Wu, X.-Y.; Zhou, J.; Jia, H.-H.; Wei, P. Extracting heavy metals from electroplating sludge by acid and bioelectrical leaching using Acidithiobacillus ferrooxidans. Hydrometallurgy 2020, 191, 105225. [Google Scholar] [CrossRef]
  265. Wang, R.; Lin, J.-Q.; Liu, X.-M.; Pang, X.; Zhang, C.-J.; Yang, C.-L.; Gao, X.-Y.; Lin, C.-M.; Li, Y.-Q.; Li, Y. Sulfur oxidation in the acidophilic autotrophic Acidithiobacillus spp. Front. Microbiol. 2019, 9, 3290. [Google Scholar] [CrossRef]
  266. Maksimova, Y.; Eliseeva, A.; Maksimov, A. Metabolic and Morphological Aspects of Adaptation of Alkaliphilic Bacillus aequororis 5-DB and Alkali-Tolerant Bacillus subtilis ATCC 6633 to Changes in pH and Mineralization. Int. J. Microbiol. 2024, 2024, 3087296. [Google Scholar] [CrossRef] [PubMed]
  267. Dixit, V.K.; Misra, S.; Mishra, S.K.; Tewari, S.K.; Joshi, N.; Chauhan, P.S. Characterization of plant growth-promoting alkalotolerant Alcaligenes and Bacillus strains for mitigating the alkaline stress in Zea mays. Antonie Leeuwenhoek 2020, 113, 889–905. [Google Scholar] [CrossRef] [PubMed]
  268. Wang, C.; Zhang, Y.; Wei, S.; Fu, L.; Wang, Y.; Jin, M. Bioconversion of soybean meal into gut microbiota-targeting polysaccharides via fermentation by Bacillus subtilis. J. Clean. Prod. 2024, 464, 142787. [Google Scholar] [CrossRef]
  269. Zampieri, B.D.B.; Nogueira, E.W.; Quaglio, O.A.; Brucha, G. Overview of known alkaliphilic bacteria from bauxite residue. J. Min. Mech. Eng. 2019, 1, 41–50. [Google Scholar] [CrossRef]
  270. Inan Bektas, K.; Nalcaoğlu, A.; Ceylan, E.; Colak, D.N.; Caglar, P.; Agirman, S.; Sivri, N.S.; Gunes, S.; Kaya, A.; Canakci, S. Isolation and characterization of detergent-compatible amylase-, protease-, lipase-, and cellulase-producing bacteria. Braz. J. Microbiol. 2023, 54, 725–737. [Google Scholar] [CrossRef]
  271. Gallo, G.; Aulitto, M. Advances in Extremophile Research: Biotechnological Applications through Isolation and Identification Techniques. Fermentation 2024, 14, 1205. [Google Scholar] [CrossRef]
  272. Schultz, J.; Modolon, F.; Peixoto, R.S.; Rosado, A.S. Shedding light on the composition of extreme microbial dark matter: Alternative approaches for culturing extremophiles. Front. Microbiol. 2023, 14, 1167718. [Google Scholar] [CrossRef] [PubMed]
  273. Vijayalaxmi, S.; Jayalakshmi, S.K.; Sreeramulu, K. Polyphenols from different agricultural residues: Extraction, identification and their antioxidant properties. J. Food Sci. Technol. 2015, 52, 2761–2769. [Google Scholar] [CrossRef]
  274. Nehmeh, M.; Rodriguez-Donis, I.; Cavaco-Soares, A.; Evon, P.; Gerbaud, V.; Thiebaud-Roux, S. Bio-Refinery of Oilseeds: Oil Extraction, Secondary Metabolites Separation towards Protein Meal Valorisation—A Review. Processes 2022, 10, 841. [Google Scholar] [CrossRef]
  275. Ancuța, P.; Sonia, A. Oil Press-Cakes and Meals Valorization through Circular Economy Approaches: A Review. Appl. Sci. 2020, 10, 7432. [Google Scholar] [CrossRef]
  276. Rakha, A.; Shehzad, A.; Khan, K. Editorial: Plant bioactives: Challenges of extraction and processing. Front. Nutr. 2024, 11, 1357925. [Google Scholar] [CrossRef] [PubMed]
  277. Sumardiono, S.; Matin, H.H.A.; Hartono, I.I.; Choiruly, L. Biogas production from corn stalk as agricultural waste containing high cellulose material by anaerobic process. Mater. Today Proc. 2022, 63, S477–S483. [Google Scholar] [CrossRef]
  278. Tajmirriahi, M.; Momayez, F.; Karimi, K. The critical impact of rice straw extractives on biogas and bioethanol production. Bioresour. Technol. 2021, 319, 124167. [Google Scholar] [CrossRef]
  279. Alavijeh, R.S.; Shahvandi, A.; Okoro, O.V.; Denayer, J.F.; Karimi, K. Biorefining of corn stover for efficient production of bioethanol, biodiesel, biomethane, and value-added byproducts. Energy Convers. Manag. 2023, 283, 116877. [Google Scholar] [CrossRef]
  280. Qian, S.; Gao, S.; Li, J.; Liu, S.; Diao, E.; Chang, W.; Liang, X.; Xie, P.; Jin, C. Effects of combined enzymatic hydrolysis and fed-batch operation on efficient improvement of ferulic acid and p-coumaric acid production from pretreated corn straws. Bioresour. Technol. 2022, 366, 128176. [Google Scholar] [CrossRef]
  281. Shankar, K.; Beladhadi, R.; Jayalakshmi, S.; Sreeramulu, K. Recovery of antioxidative phenolic compounds by the valorization of rice biomass under the influence of lignocellulolytic enzymes. Biocatal. Biotransform. 2024, 42, 440–453. [Google Scholar] [CrossRef]
  282. Oliveira, A.L.; Carvalho, M.J.; Oliveira, D.L.; Costa, E.; Pintado, M.; Madureira, A.R. Sugarcane straw polyphenols as potential food and nutraceutical ingredient. Foods 2022, 11, 4025. [Google Scholar] [CrossRef]
  283. Oleszek, M.; Kowalska, I.; Bertuzzi, T.; Oleszek, W. Phytochemicals derived from agricultural residues and their valuable properties and applications. Molecules 2023, 28, 342. [Google Scholar] [CrossRef]
  284. Risite, H.; Salim, M.H.; Oudinot, B.T.; Ablouh, E.-h.; Joyeux, H.T.; Sehaqui, H.; Razafimahatratra, J.H.A.; Qaiss, A.E.K.; El Achaby, M.; Kassab, Z. Artemisia annua stems a new sustainable source for cellulosic materials: Production and characterization of cellulose microfibers and nanocrystals. Waste Biomass Valorization 2022, 13, 2411–2423. [Google Scholar] [CrossRef]
  285. Lee, B.J.; Weyers, M.; Haynes, R.K.; van der Kooy, F. Discovery of artemisinin in Artemisia annua, its current production, and relevance to sub-Saharan Africa. S. Afr. J. Bot. 2023, 153, 21–27. [Google Scholar] [CrossRef]
  286. Cheong, D.H.; Tan, D.W.; Wong, F.W.; Tran, T. Anti-malarial drug, artemisinin and its derivatives for the treatment of respiratory diseases. Pharmacol. Res. 2020, 158, 104901. [Google Scholar] [CrossRef] [PubMed]
  287. Talib, W.H.; Alsalahat, I.; Daoud, S.; Abutayeh, R.F.; Mahmod, A.I. Plant-derived natural products in cancer research: Extraction, mechanism of action, and drug formulation. Molecules 2020, 25, 5319. [Google Scholar] [CrossRef]
  288. Xiong, X.; Gou, J.; Liao, Q.; Li, Y.; Zhou, Q.; Bi, G.; Li, C.; Du, R.; Wang, X.; Sun, T. The Taxus genome provides insights into paclitaxel biosynthesis. Nat. Plants 2021, 7, 1026–1036. [Google Scholar] [CrossRef]
  289. Yu, K.D.; Ye, F.G.; He, M.; Fan, L.; Ma, D.; Mo, M.; Wu, J.; Liu, G.Y.; Di, G.H.; Zeng, X.H.; et al. Effect of Adjuvant Paclitaxel and Carboplatin on Survival in Women with Triple-Negative Breast Cancer: A Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1390–1396. [Google Scholar] [CrossRef]
  290. Wongsirichot, P.; Gonzalez-Miquel, M.; Winterburn, J. Recent advances in rapeseed meal as alternative feedstock for industrial biotechnology. Biochem. Eng. J. 2022, 180, 108373. [Google Scholar] [CrossRef]
  291. Gołębiewska, K.; Fraś, A.; Gołębiewski, D. Rapeseed meal as a feed component in monogastric animal nutrition—A review. Ann. Anim. Sci. 2022, 22, 1163–1183. [Google Scholar] [CrossRef]
  292. Moreno-González, M.; Girish, V.; Keulen, D.; Wijngaard, H.; Lauteslager, X.; Ferreira, G.; Ottens, M. Recovery of sinapic acid from canola/rapeseed meal extracts by adsorption. Food Bioprod. Process. 2020, 120, 69–79. [Google Scholar] [CrossRef]
  293. Xie, C.; Li, W.; Gao, R.; Yan, L.; Wang, P.; Gu, Z.; Yang, R. Determination of glucosinolates in rapeseed meal and their degradation by myrosinase from rapeseed sprouts. Food Chem. 2022, 382, 132316. [Google Scholar] [CrossRef]
  294. Yang, J.; Chen, J.; Hao, Y.; Liu, Y. Identification of the DPPH radical scavenging reaction adducts of ferulic acid and sinapic acid and their structure-antioxidant activity relationship. LWT 2021, 146, 111411. [Google Scholar] [CrossRef]
  295. Kim, J.S.; Han, S.; Kim, H.; Won, S.Y.; Park, H.W.; Choi, H.; Choi, M.; Lee, M.Y.; Ha, I.J.; Lee, S.-G. Anticancer effects of high glucosinolate synthesis lines of Brassica rapa on colorectal cancer cells. Antioxidants 2022, 11, 2463. [Google Scholar] [CrossRef] [PubMed]
  296. Wilkinson, J.; Young, R. Strategies to reduce reliance on soya bean meal and palm kernel meal in livestock nutrition. J. Appl. Anim. Nutr. 2020, 8, 75–85. [Google Scholar] [CrossRef]
  297. Singhvi, M.S.; Zinjarde, S.S. Production of pharmaceutically important genistein and daidzein from soybean flour extract by using β-glucosidase derived from Penicillium janthinellum NCIM 1171. Process Biochem. 2020, 97, 183–190. [Google Scholar] [CrossRef]
  298. Berhow, M.A.; Singh, M.; Bowman, M.J.; Price, N.P.; Vaughn, S.F.; Liu, S.X. Quantitative NIR determination of isoflavone and saponin content of ground soybeans. Food Chem. 2020, 317, 126373. [Google Scholar] [CrossRef] [PubMed]
  299. Li, S.; Jin, Z.; Hu, D.; Yang, W.; Yan, Y.; Nie, X.; Lin, J.; Zhang, Q.; Gai, D.; Ji, Y. Effect of solid-state fermentation with Lactobacillus casei on the nutritional value, isoflavones, phenolic acids and antioxidant activity of whole soybean flour. LWT 2020, 125, 109264. [Google Scholar] [CrossRef]
  300. Kim, I.-S. Current perspectives on the beneficial effects of soybean isoflavones and their metabolites for humans. Antioxidants 2021, 10, 1064. [Google Scholar] [CrossRef]
  301. Aboushanab, S.A.; Khedr, S.M.; Gette, I.F.; Danilova, I.G.; Kolberg, N.A.; Ravishankar, G.A.; Ambati, R.R.; Kovaleva, E.G. Isoflavones derived from plant raw materials: Bioavailability, anti-cancer, anti-aging potentials, and microbiome modulation. Crit. Rev. Food Sci. Nutr. 2022, 63, 261–287. [Google Scholar] [CrossRef]
  302. Povod, M.; Mykhalko, O.; Povoznikov, M.; Gutyj, B.; Koberniuk, V.; Shuplyk, V.; Ievstafiieva, Y.; Buchkovska, V. Efficiency of using high-protein sunflower meal instead of soybean meal in feeding of growing piglets. Sci. Pap. Ser. Manag. Econ. Eng. Agric. Rural Dev. 2022, 22, 595–602. [Google Scholar]
  303. Bozhkov, A.A.; Ganin, V.Y.; Akzhyhitov, R.A.; Ivanov, E.G.; Bilovetska, S.G.; Dobrianska, N.I.; Novikova, A.V.; Bozhkov, A.I. Chlorogenic acid from sunflower meal regulates the number of immunocompetent cells in animals with toxic liver fibrosis. Clin. Nutr. Open Sci. 2024, 53, 78–94. [Google Scholar] [CrossRef]
  304. Ali, M.; Khalil, M.; Badawy, W.Z.; Hellwig, M. Ultrasonic treatment as a modern technique to facilitate the extraction of phenolic compounds from organic sunflower seed cakes. J. Sci. Food Agric. 2024, 104, 2245–2251. [Google Scholar] [CrossRef]
  305. Le, T.T.; Ropars, A.; Aymes, A.; Frippiat, J.-P.; Kapel, R. Multicriteria optimization of phenolic compounds capture from a sunflower protein isolate production process by-product by adsorption column and assessment of their antioxidant and anti-inflammatory effects. Foods 2021, 10, 760. [Google Scholar] [CrossRef] [PubMed]
  306. Larnaudie, V.; Ferrari, M.D.; Lareo, C. Switchgrass as an alternative biomass for ethanol production in a biorefinery: Perspectives on technology, economics and environmental sustainability. Renew. Sustain. Energy Rev. 2022, 158, 112115. [Google Scholar] [CrossRef]
  307. Bryant, N.D.; Pu, Y.; Tschaplinski, T.J.; Tuskan, G.A.; Muchero, W.; Kalluri, U.C.; Yoo, C.G.; Ragauskas, A.J. Transgenic poplar designed for biofuels. Trends Plant Sci. 2020, 25, 881–896. [Google Scholar] [CrossRef] [PubMed]
  308. Chipkar, S.H. Studying Effect of Drought on Switchgrass and Identifying Associated Microbial Inhibitors. Master’s Thesis, Michigan Technological University, Houghton, MI, USA, 2023. [Google Scholar]
  309. Devi, M.; Bamrah, P.K.; Goyal, R.; Choudhary, M.; Chopra, H. Insights on the emerging therapeutic potential of terpenoids as anti-inflammatory agents: A scoping review. J. Bio-X Res. 2024, 7, 0006. [Google Scholar] [CrossRef]
  310. De Paola, C. Enhancing Nicotiana benthamiana as Chassis for Molecular Farming: Targeting Flowering Time for Increased Biomass and Recombinant Protein Production. Ph.D. Thesis, Universitat Politècnica de València, Valencia, Spain, 2024. [Google Scholar]
  311. Naeem, M.; Han, R.; Ahmad, N.; Zhao, W.; Zhao, L. Tobacco as green bioreactor for therapeutic protein production: Latest breakthroughs and optimization strategies. Plant Growth Regul. 2024, 103, 227–241. [Google Scholar] [CrossRef]
  312. Wu, S.; Gao, C.; Pan, H.; Wei, K.; Li, D.; Cai, K.; Zhang, H. Advancements in tobacco (Nicotiana tabacum L.) seed oils for biodiesel production. Front. Chem 2022, 9, 834936. [Google Scholar] [CrossRef]
  313. Botero Gutiérrez, C.D.; Restrepo Serna, D.L.; Cardona Alzate, C.A. A comprehensive review on the implementation of the biorefinery concept in biodiesel production plants. Biofuel Res. J. 2017, 4, 691–703. [Google Scholar] [CrossRef]
  314. Kumar, L.; Mohan, L.; Anand, R.; Joshi, V.; Chugh, M.; Bharadvaja, N. A review on unit operations, challenges, opportunities, and strategies to improve algal based biodiesel and biorefinery. Front. Chem. Eng. 2022, 4, 998289. [Google Scholar] [CrossRef]
  315. Makepa, D.; Chihobo, C. Barriers to commercial deployment of biorefineries: A multi-faceted review of obstacles across the innovation chain. Heliyon 2024, 10, e32649. [Google Scholar] [CrossRef]
  316. Seufitelli, G.V.S.; El-Husseini, H.; Pascoli, D.U.; Bura, R.; Gustafson, R. Techno-economic analysis of an integrated biorefinery to convert poplar into jet fuel, xylitol, and formic acid. Biotechnol. Biofuels Bioprod. 2022, 15, 143. [Google Scholar] [CrossRef]
  317. Biorefinery Demo Plant—Sekab. Available online: https://www.sekab.com/en/this-is-how-it-works/biorefinery-demo-plant/ (accessed on 8 May 2025).
  318. Borregaard—The Sustainable Biorefinery. Available online: https://www.borregaard.com/ (accessed on 8 May 2025).
  319. Integrating the Production of Biofuels and Bioproducts. Available online: https://www.energy.gov/eere/articles/integrating-production-biofuels-and-bioproducts/ (accessed on 8 May 2025).
  320. Qiao, J.; Cui, H.; Wang, M.; Fu, X.; Wang, X.; Li, X.; Huang, H. Integrated biorefinery approaches for the industrialization of cellulosic ethanol fuel. Bioresour. Technol. 2022, 360, 127516. [Google Scholar] [CrossRef] [PubMed]
  321. Vlysidis, A.; Binns, M.; Webb, C.; Theodoropoulos, C. A techno-economic analysis of biodiesel biorefineries: Assessment of integrated designs for the co-production of fuels and chemicals. Energy 2011, 36, 4671–4683. [Google Scholar] [CrossRef]
  322. Keiper, F.; Atanassova, A. Regulation of Synthetic Biology: Developments Under the Convention on Biological Diversity and Its Protocols. Front. Bioeng. Biotechnol. 2020, 8, 310. [Google Scholar] [CrossRef] [PubMed]
  323. Naveen, A.K.; Sontakke, M. A review on regulatory aspects, challenges and public perception in acceptance of genetically modified foods. Food Sci. Biotechnol. 2024, 33, 791–804. [Google Scholar] [CrossRef]
  324. Patraşcu, I.; Bîldea, C.S.; Kiss, A.A. Eco-efficient Downstream Processing of Biobutanol by Enhanced Process Intensification and Integration. ACS Sustain. Chem. Eng. 2018, 6, 5452–5461. [Google Scholar] [CrossRef]
  325. Tönjes, S.; Uitterhaegen, E.; Palmans, I.; Ibach, B.; De Winter, K.; Van Dijck, P.; Soetaert, W.; Vandecruys, P. Metabolic Engineering and Process Intensification for Muconic Acid Production Using Saccharomyces cerevisiae. Int. J. Mol. Sci. 2024, 25, 10245. [Google Scholar] [CrossRef]
  326. Biological Approvals by Year. Available online: https://www.fda.gov/vaccines-blood-biologics/development-approval-process-cber/biological-approvals-year (accessed on 26 May 2025).
  327. Kohli, K.; Prajapati, R.; Sharma, B.K. Bio-Based Chemicals from Renewable Biomass for Integrated Biorefineries. Energies 2019, 12, 233. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of integrated biorefinery strategies for co-producing biofuels and pharmaceuticals.
Figure 1. Schematic overview of integrated biorefinery strategies for co-producing biofuels and pharmaceuticals.
Fermentation 11 00312 g001
Figure 2. Critical future research directions for integrated biofuel–pharmaceutical co-production systems.
Figure 2. Critical future research directions for integrated biofuel–pharmaceutical co-production systems.
Fermentation 11 00312 g002
Table 1. Examples of microbial metabolites for dual biofuel and pharmaceutical production.
Table 1. Examples of microbial metabolites for dual biofuel and pharmaceutical production.
MicroorganismBiofuel(s)/PrecursorsPharmaceuticals/PrecursorsRefs.
Acinetobacter spp.BiodieselPotential for emulsifiers/biosurfactants—varies by species[17,18]
Bacillus spp.BiodieselBioacetoin[19,20]
Chlamydomonas reinhardtiiHydrogen, BiodieselCarotenoids (β-carotene, astaxanthin)[21,22,23]
Clostridium acetobutylicumButanolButyric Acid[24,25]
Dunaliella salinaBiodieselβ-carotene (antioxidant, vitamin A precursor)[26,27,28]
Haematococcus pluvialisBiodiesel, Ethanol, Biogas (from residual biomass)Astaxanthin (antioxidant, anti-inflammatory, neuroprotective)[29,30,31]
Mortierella isabellinaBiodieselGamma-linolenic acid (anti-inflammatory)[32,33,34]
Phaffia rhodozymaBiodiesel (from lipids)Astaxanthin (antioxidant, anti-inflammatory, neuroprotective)[35,36]
Phormidium sp.BiodieselSqualene[37,38]
Rhizopus oryzaeBiodieselChitin/Chitosan (biomedical polymers)[39,40,41]
Rhodotorula babjevae Y-SL7Microbial oil (>40%)Polyol Esters of Fatty Acids [42,43]
Rhodotorula glutinisBiodiesel Carotenoids (β-carotene, torulene, torularhodin-antioxidants)[44,45]
Rhodotorula kratochvilovae SY89BiodieselCarotenoids, β-glucans (immunostimulatory), Exoglycolipids[46,47,48]
Rhodococcus spp.BiodieselPotential for steroid biotransformation—varies by species[49,50,51]
Spirulina (Arthrospira platensis)BiodieselPhycocyanin (antioxidant, anti-inflammatory)[52,53]
Yarrowia lipolyticaBiodieselOmega-3 fatty acids, Carotenoids, Flavonoids (engineered, e.g., naringenin), Lipases[54,55,56,57,58,59]
Table 2. Examples of synthetic biology platforms for dual applications.
Table 2. Examples of synthetic biology platforms for dual applications.
Engineered OrganismSynthetic Biology ApproachBiofuel(s)/PrecursorsPharmaceuticalsRefs.
Escherichia coliPathway engineering, Gene expression optimization, Metabolic flux controlFatty acid ethyl esters, Alkanes, Isoprenoid fuelsLycopene, Amorphadiene (artemisinin precursor), Other isoprenoids[82,83,84,85,86,87,88]
Microbial ConsortiaDivision of labor, Cross-feeding between engineered strainsVaries (e.g., H2 + CO2 -> methane/acetate)Varies (e.g., sequential biotransformation for complex drugs)[89,90,91,92]
Saccharomyces cerevisiaeMetabolic engineering, Heterologous pathway construction, CRISPR-Cas9Ethanol, Isobutanol, Farnesene, Other alcoholsArtemisinic acid (artemisinin precursor), Resveratrol, Insulin precursors, Polyketides[93,94,95,96,97,98,99]
Synechocystis sp. PCC 6803 (Cyanobacterium)Photosynthetic pathway engineering, CO2 fixation enhancement, CRISPR-Cas9Isobutanol, Ethanol, Fatty acids, Limoneneβ-carotene, Phycocyanin, Terpenoid precursors, Other bioactive compounds[100,101,102,103,104,105]
Various (using advanced tools)Biosensors, Dynamic regulatory circuits, High-throughput screeningOptimized yield/titerOptimized yield/titer, Balanced co-production[106,107,108]
Yarrowia lipolyticaMetabolic engineering, Lipid pathway optimization, CRISPR-Cas9, Pathway compartmentalizationBiodiesel Omega-3 fatty acids (EPA, DHA), Carotenoids, Polyketides[59,109,110,111,112,113,114,115]
Table 3. Examples of medicinal components extracted from biorefinery waste.
Table 3. Examples of medicinal components extracted from biorefinery waste.
Biorefinery Waste StreamExtracted Medicinal Component(s)Potential Medicinal Application(s)Refs.
Crude Glycerol from BiodieselImpurities potentially convertible, e.g., to 1,3-propanediolChemical intermediate[122,123,124]
Fermentation ResiduesResidual enzymes, Bioactive Peptides, Phenolic compounds, Microbial biomass componentsAntimicrobial, Antioxidant, Enzyme source, Animal feed supplement[125,126,127,128,129]
Fungal Biomass ResiduesChitin, ChitosanDrug delivery, Wound healing, Tissue engineering[130,131,132,133,134,135]
Hemicellulose HydrolysatePhenolic acids, OligosaccharidesAntioxidant, Prebiotic[136,137,138,139]
Lignocellulosic ResiduesPhenolic monomers/oligomersAntioxidant, Antimicrobial, Anti-inflammatory, Flavorant, Chemical precursor[140,141,142,143,144,145,146,147]
Spent Microalgal Biomass (post-primary extraction)Bioactive Peptides, Proteins, β-glucans, Residual Carotenoids, Chlorophyll derivatives, Omega-3 fatty acids, MineralsAntioxidant, Antihypertensive, Immunostimulatory, Antimicrobial, Nutritional supplement[145,148,149,150,151,152]
Table 4. Examples of cross-disciplinary applications of nanocatalysts.
Table 4. Examples of cross-disciplinary applications of nanocatalysts.
Nanocatalyst TypeProcess/Reaction CatalyzedRelevance to Co-productionRefs.
Enzyme–Nanomaterial ConjugatesTransesterification; Esterification; Hydrolysis; Kinetic ResolutionEfficient biodiesel production; Synthesis/resolution of chiral drug precursors; Catalyst recovery and reuse via magnetic/physical separation.[160,161,162,163,164]
Supported Metal NPsHydrogenation, Dehydrogenation, Oxidation, C-C Coupling, HydrodeoxygenationUpgrading biomass platform chemicals into fuel additives and pharmaceutical building blocks; Lignin depolymerization.[165,166,167,168]
Metal Oxide NanocatalystsPhotocatalysis; Oxidation; Acid/Base CatalysisBiomass pretreatment enhancement; Biorefinery wastewater treatment; Specific organic synthesis for pharmaceuticals; Support material.[169,170,171,172,173,174]
Bimetallic/Alloy NanoparticlesSynergistic CatalysisEnhanced activity/selectivity for hydrogenation, oxidation, C-C coupling in biomass upgrading, potentially benefiting both fuel/pharma pathways.[175,176,177,178,179]
Nanocatalysts for Lignin ValorizationSelective Cleavage of Lignin BondsControlled depolymerization of lignin waste into specific aromatic precursors for high-performance biofuels/materials and pharmaceuticals.[180,181,182,183]
Multifunctional/Tandem NanocatalystsCascade ReactionsOne-pot conversion of biomass components or intermediates into advanced biofuels and pharmaceutical precursors, improving process efficiency.[184,185,186,187,188]
Table 5. Examples of computer-aided design applications in co-production optimization.
Table 5. Examples of computer-aided design applications in co-production optimization.
Computational Tool/MethodBiological System/Process TargetedRole in Optimizing Co-productionRefs.
Genome-Scale Metabolic Models (GEMs)Whole-cell metabolism (bacteria, yeast, algae, etc.)Comprehensive representation of metabolic network; Foundation for constraint-based modeling and systems analysis.[204,205,206,207,208,209]
Flux Balance Analysis (FBA) and Variants (OptFlux, FVA)Microbial metabolic flux distributionPredicting optimal flux patterns; Identifying gene modification targets (knockouts, overexpression) for redirecting flux towards simultaneous biofuel/pharma production.[210,211,212,213]
13C-Metabolic Flux AnalysisIntracellular metabolic fluxesQuantifying actual metabolic fluxes to validate and refine GEMs and FBA predictions for co-production pathways.[214,215,216]
Pathway Design Algorithms (e.g., RetroPath, BNICE)Novel metabolic pathway constructionDesigning synthetic pathways for target molecules; Exploring alternative routes; Identifying pathways potentially favoring co-production economics.[217,218,219]
Molecular Dynamics SimulationEnzyme structure function, Enzyme–substrate interactionsUnderstanding enzyme mechanisms; Guiding protein engineering efforts to improve catalyst efficiency or specificity relevant to biofuel/pharma synthesis.[220,221,222,223]
Process Simulation Software (e.g., Aspen, SuperPro)Integrated biorefinery processes (upstream, fermentation, downstream)Designing, optimizing, and scaling up entire processes; Performing techno-economic analysis and life cycle assessment of co-production scenarios.[224,225,226]
Machine Learning (ML)/Artificial Intelligence (AI)Multi-omics data, Fermentation data, Genetic circuit designBuilding predictive models from complex data; Optimizing conditions; Discovering enzyme functions; Designing dynamic control systems for balancing co-production.[227,228,229,230,231]
Table 6. Examples of extremophile co-development for dual applications.
Table 6. Examples of extremophile co-development for dual applications.
Extremophile Type/ExampleExtreme Condition ToleranceBiofuel Product/Process ContributionPharmaceutical/Value-Added Product/EnzymeRefs.
Thermophilic Bacteria/Archaea (Clostridium, Caldicellulosiruptor, Thermoanaerobacterium)High Temperature (>60 °C)Ethanol, Hydrogen, Butanol (from lignocellulose via CBP); Reduced contaminationThermostable enzymes (cellulases, xylanases, ligninases, amylases, proteases)[194,241,242,243,244]
Halophili Algae (Dunaliella salina)High Salinity (>10–30% NaCl)Lipids (biodiesel precursor), Glycerolβ-carotene, Lutein[26,245,246,247,248,249,250]
Halophilic Archaea (Halobacterium, Haloferax)High Salinity (15%—saturation)Lipids (potential), PHAs (bioplastics/biomedical), Gas vesicles (potential)Bacterioruberin (C50 carotenoid antioxidant), Compatible solutes (ectoine, hydroxyectoine)[251,252,253,254,255,256,257]
Psychrophilic Bacteria/Algae/FungiLow Temperature (<15 °C)Lipids (biodiesel precursor-potential)Polyunsaturated fatty acids, Cold-active enzymes (lipases, proteases, amylases)[258,259,260,261]
Acidophilic Microorganisms (Acidithiobacillus)Low pH (<3)Biomining contribution (metal leaching-indirect)Acid-stable enzymes, Unique metabolites[262,263,264,265]
Alkaliphilic Microorganisms (Bacillus spp.)High pH (>9)Contribution to specific bioconversionsAlkali-stable enzymes (proteases, amylases for detergents), Unique metabolites[266,267,268,269,270]
Table 7. Examples of integrated utilization of plant secondary metabolites.
Table 7. Examples of integrated utilization of plant secondary metabolites.
Plant Source/TypePrimary Biomass Use (Biofuel Potential)Secondary Metabolite(s)Medicinal Application(s)/Potential BenefitsRefs.
Crop Residues (Corn Stover, Wheat Straw)Cellulosic Ethanol, BiogasPhenolic Acids, FlavonoidsAntioxidant, Anti-inflammatory[277,278,279,280,281,282,283]
Artemisia annuaLignocellulosic Biomass Residue (Post-extraction)ArtemisininAntimalarial[284,285,286]
Taxus spp.Lignocellulosic Biomass Residue (Needles/Bark, Post-extraction)PaclitaxelAnticancer[287,288,289]
Rapeseed Meal (Post-oil extraction)Animal Feed/Fertilizer/Potential Energy RecoveryPhenolic Compounds, GlucosinolatesAntioxidant, Potential anticancer (glucosinolate metabolites)[290,291,292,293,294,295]
Soybean Meal (Post-oil extraction)Animal Feed/Fertilizer/Potential Energy RecoveryIsoflavones, Saponins, Phenolic acidsPhytoestrogenic (hormone health), Antioxidant, Potential anticancer[296,297,298,299,300,301]
Sunflower Meal (Post-oil extraction)Animal Feed/Fertilizer/Potential Energy RecoveryChlorogenic acid, Other phenolic compoundsAntioxidant, Anti-inflammatory[302,303,304,305]
Dedicated Energy Crops (Switchgrass, Poplar)Cellulosic BiofuelsPhenolics, Flavonoids, Terpenoids—variesVaries (Antioxidant, Anti-inflammatory potential, requires investigation)[306,307,308,309]
Tobacco (Nicotiana spp.—engineered)Potential for Oil (biodiesel)/BiomassEngineered production of specific proteins/antibodiesTherapeutic proteins (e.g., vaccines, antibodies)[310,311,312]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liu, T.; He, M.; Shi, R.; Yin, H.; Luo, W. Biofuel–Pharmaceutical Co-Production in Integrated Biorefineries: Strategies, Challenges, and Sustainability. Fermentation 2025, 11, 312. https://doi.org/10.3390/fermentation11060312

AMA Style

Liu T, He M, Shi R, Yin H, Luo W. Biofuel–Pharmaceutical Co-Production in Integrated Biorefineries: Strategies, Challenges, and Sustainability. Fermentation. 2025; 11(6):312. https://doi.org/10.3390/fermentation11060312

Chicago/Turabian Style

Liu, Tao, Miaoxin He, Rui Shi, Hui Yin, and Wen Luo. 2025. "Biofuel–Pharmaceutical Co-Production in Integrated Biorefineries: Strategies, Challenges, and Sustainability" Fermentation 11, no. 6: 312. https://doi.org/10.3390/fermentation11060312

APA Style

Liu, T., He, M., Shi, R., Yin, H., & Luo, W. (2025). Biofuel–Pharmaceutical Co-Production in Integrated Biorefineries: Strategies, Challenges, and Sustainability. Fermentation, 11(6), 312. https://doi.org/10.3390/fermentation11060312

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop