Skip to main content
3 Biotech logoLink to 3 Biotech
. 2024 Mar 18;14(4):112. doi: 10.1007/s13205-024-03958-z

Therapeutic proteins: developments, progress, challenges, and future perspectives

Vimal Kumar 1, Arti Barwal 2, Nitin Sharma 3, Danish Shafi Mir 1, Pradeep Kumar 4, Vikas Kumar 1,
PMCID: PMC10948735  PMID: 38510462

Abstract

Proteins are considered magic molecules due to their enormous applications in the health sector. Over the past few decades, therapeutic proteins have emerged as a promising treatment option for various diseases, particularly cancer, cardiovascular disease, diabetes, and others. The formulation of protein-based therapies is a major area of research, however, a few factors still hinder the large-scale production of these therapeutic products, such as stability, heterogenicity, immunogenicity, high cost of production, etc. This review provides comprehensive information on various sources and production of therapeutic proteins. The review also summarizes the challenges currently faced by scientists while developing protein-based therapeutics, along with possible solutions. It can be concluded that these proteins can be used in combination with small molecular drugs to give synergistic benefits in the future.

Keywords: Therapeutic proteins, Diseases, Sources, Production, Challenges

Introduction

Protein-based treatments remain a top priority for many pharmaceutical businesses in the modern era of drug discovery and development (R&D). Recombinant biologics have been widely employed as the medicine of choice for treating a variety of diseases, in large part due to their specificity in identifying and modifying disease-related pathways (Walsh 2018). Human insulin was the first recombinant therapeutic protein, and its discovery in 1970 has increased interest in and expectations for the creation of new protein-based medicines. The 1st therapeutic protein other than antibodies, Insulin, was purified from the animal pancreas (bovine and porcine pancreas) and administered to patients suffering from diabetes mellitus type I (DM‑I) and type II (DM-II) in 1922. It was the first paradigm shift. DM-I is a disease whose primary cause is the absence of the protein hormone insulin, which provides signals to the cells so that the cells can perform functions related to glucose metabolism. However, its use was limited due to its unavailability, cost, and immunogenicity. Then, it took 60 years for the second paradigm to produce the first recombinant therapeutic protein, i.e., humulin (human insulin), with the application of genetic engineering, which was approved by the US FDA in 1982. Later, a murine mAB Muromonab-CD3 (OKT3) was the first therapeutic protein approved for clinical use in 1986, but it was withdrawn from the market in 2010.

Protein-based therapeutics are the fastest-growing medicines in the pharmacological industry (Lagassé et al. 2017). Most protein therapeutics are recombinants, and more than 170 are used in medicine worldwide. In contrast, many of them are in clinical trials used for multiple applications such as diagnosis, prevention therapy of cancers, immune disorders, infections, and disease management. They have become an essential part of the pharmaceutical industry, and the pharma industries are focused on research using these proteins to identify novel effective therapeutics (Kintzing et al. 2016). These therapeutics have been known to represent 1/3rd of the entities launched as new medicines under biology. More than 6 million people have lost their lives due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), making the outbreak the deadliest global health crisis since the influenza pandemic of 1918 (Aleem et al. 2023). Human recombinant soluble ACE2 protein is also one of the proposed COVID-19 treatments. This Recombinant human-soluble ACE2 protein prevents the SARS-CoV-2 from binding to ACE2 (Zhou et al. 2020; Monteil et al. 2020; Tai et al. 2020). Additionally, peptide-based therapies were developed and evaluated to target COVID-19, focusing on the N-protein and cytolytic T-cell response (Amawi et al. 2020). Neutralizing antibodies were also evaluated for treating COVID-19 infection using similar mechanisms, which involve inhibiting virus particle binding with ACE2 receptors (Du et al. 2021).

According to La Merie's financial reports, the global value of therapeutic biologics increased annually, reaching an estimated $800 billion between 2014 and 2018 (Walsh 2018). The global market for therapeutic proteins has been proliferating in recent years, with estimates suggesting a compound annual growth rate of 8.5% between 2020 and 2027 (Research and Markets, 2021). In 2020, the market size was valued at USD 168.5 billion, with North America and Europe accounting for the largest share of the market. The therapeutic protein market in India is also growing, driven by factors such as a large patient population, rising disposable incomes, and the increasing prevalence of chronic diseases. According to a report by Grand View Research (2021), the Indian market for therapeutic proteins was valued at USD 1.34 billion in 2020 and is expected to grow at a compound annual growth rate of 8.8% between 2021 and 2028. Therapeutic proteins represent a significant and growing market globally and in India, with potential for further growth as new drugs are developed and personalized medicine becomes more widespread. Drug discovery and development have shifted their focus to therapeutic proteins (with improved human safety profiles), but they still face some obstacles. Immune responses and other side effects have been linked to various protein-based therapies.

The field of peptide drug discovery has demonstrated significant dynamism in recent times such as the development of new peptide-focused industries and the emergence of big-pharma business (Albericio and Kruger 2012; Henninot et al. 2018; Al Shaer et al. 2020; De La Torre and Albericio 2020). Therapeutic peptides are a distinct class of pharmaceutical drugs made of well-arranged sequences of amino acids, most of which have molecular weights between 500 and 5000Da (Henninot et al. 2018). Peptides are known to be extremely effective and selective while also being quite safe and well-tolerated. With over 7000 naturally occurring peptides known, many have important functions in human physiology, acting as growth factors, hormones, neurotransmitters, ion channel ligands, or anti-infectives, among other things (Giordano et al. 2014; Robinson et al. 2014; Padhi et al. 2014; Buchwald et al. 2014; Fosgerau and Hoffmann 2015). Peptides are highly effective and selective signaling molecules that attach to particular cell surface receptors, including ion channels or G protein-coupled receptors (GPCRs), to initiate intracellular actions. The production complexity of peptide therapeutics is generally lower than that of protein-based biopharmaceuticals. Consequently, the production costs of peptide therapeutics are also less than those of small molecules (Fosgerau and Hoffmann 2015). Naturally occurring peptides (e.g., cyclosporin, oxytocin and insulin (Marx 2005; Tsomaia 2015)) due to their shortcoming such as poor physical and chemical stability as well as short half-life period in plasma circulation also a big challenge. In recent times such shortcomings are resolved using models such as “traditional design” (e.g., SWOT analysis). In addition to traditional peptide designs, a variety of peptide technologies have emerged such as peptide drug conjugates, multifunctional and cell penetrating peptides along with alternate routes of drug administration, signifying the potential and future possibilities in the peptide domain (Fosgerau and Hoffmann 2015).

Peptides mediate numerous vital biological activities, including signal transduction, heart rate regulation, food intake, and growth. They are distinguishable from proteins by their smaller size (50 amino acids or less). Peptides have a strong and selective ability to bind large protein targets, which means they are less likely to cause toxicity and off-target adverse effects than small molecule medications (Craik et al. 2013; Tsomaia 2015; Wang et al. 2022). In contrast to small molecules that might have adverse effects by generating toxic metabolites that build up in various organs, peptides break down into amino acids, hence reducing the potential for toxicity (Manna et al. 2018; Wang et al. 2022). Peptides are more stable at room temperature, have a cheaper manufacturing cost, and have a greater capacity to penetrate tissues because of their smaller size than recombinantly generated antibodies and designed protein medicines (Patel et al. 2019; Lee et al. 2019; Barman et al. 2023). Furthermore, a peptide sequence can be edited to include non-natural building blocks and other chemical scaffolds, resulting in a palette of modified peptides with a variety of functionalities and chemical diversity (Oeller et al. 2023). With the dawn of the twenty-first century, peptide drug development entered a new age marked by a major acceleration in structural biology, recombinant biologics and new synthetic and new analytical technologies (Wang et al. 2022). Currently, metabolic illnesses and oncology are the two main disease categories that are driving the therapeutic usage of peptide medicines. There are now around 170 peptides undergoing clinical trials, and numerous others are in preclinical development. Table 1 shows some of the FDA approved (2015–2020) therapeutic peptide drugs in clinical use and for management of various diseases.

Table 1.

List of some FDA approved (2015–2020) therapeutic peptide drugs in clinical use and for management of various diseases

S. no Therapeutic peptide Target name Indications Year of approval References
1 Setmelanotide Melanocortin-4 receptor For chronic weight management of obesity 2020 Kamermans et al. (2019), Haws et al. (2020), Wang et al. (2022)
2 Edotreotide gallium Ga-68 Somatostatin receptors Used to identify neuroendocrine tumors that are positive for the somatostatin receptor 2019 Moyade and Vinjamuri (2019, Evangelista et al. (2020)
3 Lutetium Lu 177 dotatate Somatostatin receptors In treatment of gastroenteropancreatic neuroendocrine tumors 2018 Das et al. (2019), Kaewput and Vinjamuri (2022)
4 Etelcalcetide CaSR In treatment of secondary hyperparathyroidism 2016 Friedl and Zitt (2018)
5 Angiotensis II AT1 Receptor In treatment of sepsis and septic shock 2017 Fernandes et al. (2021)
6 Insulin degludec Tresiba® - In treatment of diabetes 2015 Kesavadev et al. (2022)
7 Bremelanotide MC receptors In treatment of hypoactive sexual desire disorder 2019 Rao and Andrade (2020)
8 Plecanatide Guanylate cyclase C In treatment of idiopathic constipation 2017 Sharma et al. (2019)
9 Macimoreline Macrilen® Ghrelin receptor (7TM receptor) In treatment of Growth Hormone deficiency 2017 Garcia et al. (2021); Yuen et al. (2023)
10 Semaglutide GLP-1 receptor In treatment of Type-2 diabetes mellitus 2017 Pearson et al. (2019)

In the current review, we offer detailed insights into the classification of therapeutic proteins and various fundamental factors that may play an essential role in the production of therapeutic proteins and may influence their production, formulation, or delivery as drugs. Moreover, we have also addressed the challenges associated with the production of therapeutic proteins.

Classification of therapeutic proteins:

Therapeutic proteins have been categorized into four groups based on their function and therapeutic applications (Leader et al. 2008; Naz et al. 2022). Group I and II are FDA-approved, while Group III and IV were investigated in vivo or in vitro. Group I includes therapeutic proteins having enzymatic or regulatory activity. It is further sub-divided into three sub-groups: 1a, Ib, and Ic. Group II includes therapeutic proteins with unique targeting activity. It is divided into two sub-groups: IIa and IIb. Group III consists of those proteins that act as prophylactic and therapeutic vaccines. Hepatitis B surface Ag (HBsAg) for Hepatitis B infection and anti-rhesus (rh) IgG vaccine, used in routine postpartum prevention of Rh(D) immunization in Rh(D) negative women (Crosnier et al. 1981; MacKenzie et al. 2004) are the examples of Group III therapeutic proteins. Group IV consists of proteins used in disease diagnostics, such as infectious disease detection and cancer detection as an imaging agent (Sodee et al. 2000; Campos-Neto et al. 2001). Table 2 summarizes the types of therapeutic proteins, their sub-groups, their function, and examples.

Table 2.

Classification of therapeutic proteins, along with functions and examples

Group Sub-group Functions Examples References
Group-I Ia

Replace a protein that is deficient or abnormal

Treat metabolic disorders or endocrine dysfunctions such as IL-1 Ra for type 2 diabetes mellitus and disorders like haemophilia A

Factor VIII (Hemophilia A) Roth et al. (2001), Akash et al. (2012b, a)
Ib Stimulate various haematological and body responses

INF-α (Hepatitis C)

Erythropoietin (chronic anaemia)

Corwin et al. (2002), van Zonneveld et al. (2004)
Ic Modify the pathophysiology of the disease

Botulinum toxin type A and B (dystonia)

Lepirudin (heparin-induced thrombocytopenia)

Oates et al. (1991), Eriksson et al. (1997)
Group-II IIa —Either activate a signalling pathway or impair their ability to function

• Cetuximab-Monoclonal antibody that binds to EGFR to treat colorectal, neck and head cancer

• Anakinra (IL-1Ra)

Saltz et al. (2004)
IIb —Can deliver proteins specifically and on-target • Ibritumomab tiuxetan, denileukin diftitox for treating non-Hodgkin’s lymphoma and persistent cutaneous T-Cell lymphoma Witzig et al. (2002), Ohya and Matsuda (2005)
Group-III IIIa Protecting against a harmful foreign agent

• Hepatitis B surface Ag (HBsAg) for Hepatitis B infection

• Anti-rhesus (rh) IgG vaccine, used in routine postpartum prevention of Rh (D) immunization in Rh(D) negative women

Crosnier et al. (1981), MacKenzie et al. (2004)
IIIb Treating an autonomous disease
IIIc Treating cancer
Group-IV Group IV proteins are not used to treat disease, but they are mentioned here because they are crucial in the decision-making process before treating and managing many diseases. These proteins are purified, and recombinant proteins are used for medical diagnostics (both in vivo and in vitro)

• Recombinant purified protein derivatives (DPPD) for diagnosis of tuberculosis exposure

• Capromab pendetide for Prostate cancer detection

• Apcitide for Imaging of acute venous thrombosis

Duchin et al. (1997), Coler et al. (2000), Sodee et al. (2000), Campos-Neto et al. (2001)

Sources of therapeutic proteins

Producing therapeutic proteins is one of the fastest-growing areas of molecular medicine, and it plays an important role in preventing and curing several disorders. Various sources of therapeutic proteins have been discussed below:

Microbes as a source of therapeutic proteins

Since nature is an attractive source of therapeutic products, huge chemical diversity is found in millions of species of plants, animals, marine organisms, and especially microorganisms. These microbes have been used for years for the large-scale production of various therapeutic products. They play an essential role as host expression systems (prokaryotic and eukaryotic) in having potential as natural sources of drugs for the treatment and prevention of several diseases such as Crohn’s disease, diarrhea, diabetes, cancer, atopic dermatitis, ulcerative colitis, etc. (Kota et al. 2018). Depending on their ability to produce the desired chemical, prokaryotic and eukaryotic microorganisms have been utilized as hosts for producing therapeutic proteins. Prokaryotes generally express smaller proteins and eukaryotes express larger ones due to their different cellular structures. Prokaryotic systems such as E. coli are used for the cost-effective, easiest, and fastest expression of therapeutic proteins. In contrast, insect systems, mammalian cells and fungi are better for the proteins requiring glycosylation (Schillberg et al. 2019). Yeasts can give higher yields of native glycosylated proteins (~ 50 kDa) at lower costs (Demain and Vaishnav 2009). Mammalian cell lines are most frequently used to assemble recombinant mammalian proteins. Genetically modified animals can also be exploited to produce therapeutic proteins, which they may secrete in their blood, urine, or milk (Jazayeri et al. 2018; Hunter et al. 2019).

Bacteria

E. coli is the earliest and most preferred microorganism employed for the expression of heterologous proteins, accounting for around 30% of the overall production of recombinant proteins in the past (Terpe 2006; Ferrer-Miralles et al. 2009). Due to improved genetic tools and a better understanding of its transcriptional and translational machinery, E. coli has been used as an expression host for large-scale production of proteins, particularly non-glycosylated proteins (Terpe 2006; Baeshen et al. 2015; Naz et al. 2022). The lac, tac, trc promoters of E. coli can be utilized to enhance protein productivity using simple and inexpensive media ingredients (Maksum et al. 2020). Solubilization of bacterial therapeutic proteins can be improved using denaturants and reducing agents to eliminate S–S bonds (Baeshen et al. 2015). Gram-positive Bacilli bacteria can also be employed for heterologous/homologous enzyme expression. Recombinant proteins are expressed in their native form using Bacillus sp. Furthermore, it has improved growth characteristics and a robust metabolism without producing exo- or endotoxins, resulting in the cost-effectiveness of a therapeutic peptide. Protein secretion into the medium eliminates the need for cell disruption and chemical processing and assists in downstream processing. Although the protein outputs from Bacillus are quite high, the production of proteases may degrade the recombinant protein (Pero and Sloma 1993). Neutral protease (metalloprotease) and Subtilisin (serine protease) account for approximately 96–98% of extracellular protease activity. So far, six to eight extracellular proteases have been identified. Murashima et al. (2002) (Murashima et al. 2002) had developed a strain lacking these eight extracellular proteases using genetic engineering techniques. Bacillus licheniformis, an exo-protease-deficient host strain, is utilized specifically for heterologous protein expression (Cai et al. 2017). Bacillus brevis is beneficial for manufacturing therapeutic proteins because it produces proteinase inhibitors (Sagiya et al. 1994; Udaka and Yamagata 2020). However, bacteria frequently cannot synthesize a recombinant human protein of the wild type due to its simple expression machinery. Bacterial cells are not developed with post-translational modifications (PTM), a feature of higher organisms, and glycosylation cannot be fully accomplished in bacterial cells. However, the low cost and simplicity of culturing bacteria are two advantages of bacterial cells over others, making them the preferred choice for lab and industrial-scale production (Kamionka 2011). But limitations of bacterial expression systems, such as lack of PTM and glycosylation, eukaryotic expression systems, such as yeast cells, fungal cells, and algal cells, were developed for therapeutic protein production (Fahad et al. 2015).

Yeasts

Yeasts are microscopic unicellular eukaryotic cells which play an important role being a good expression system for the production of therapeutic proteins because of their rapid growth rate, ease for genetic manipulation, less expensive growth medium requirements, availability of entire genome sequences of yeast and ability to provide PTMs (Nielsen 2013; Fletcher et al. 2016; Kim and Kim 2017; Vieira Gomes et al. 2018; Baghban et al. 2019; Huertas and Michán 2019). Pichia pastoris (non-conventional yeast) and Saccharomyces cerevisiae (traditional baker’s yeast) is the most used expression host systems (Love et al. 2018); however, Hansenula polymorpha and Yarrowia lipolytica have also been in use. They can readily accommodate genetic modifications, grow in simple media, and incorporate post-transformational modifications. Yeasts are effective at producing insulin, glargine, and other monoclonal antibodies (mAbs) (Mengdai et al. 2020). S. cerevisiae has several benefits over bacteria as a cloning host, including the ability to carry glycosylation and secrete heterologous proteins into the medium by incorporating signal sequences. However, glycosylation using S. cerevisiae is prohibited for mammalian proteins because O-linked oligosaccharides contain only mannose (Gellissen et al. 1992). This yeast may also induce immunological issues, as well as a decrease in activity and receptor binding at glycosylated N-linked regions. Urate oxidase, granulocyte–macrophage colony-stimulating factor (GM-CSF), insulin, glucagon, platelet-derived growth factor, hepatitis B surface antigen, and hirudin are all therapeutic compounds produced by S. cerevisiae (Gellissen et al. 1992). Recombinant yeast, mould, mammalian cells, or insects can glycosylate human chorionic gonadotropin (HCG) or erythropoietin (Saul and Sudbery 1985). Additional carbohydrates related to the oxygen moiety of threonine or serine are found in fungal therapeutic proteins (Nunberg et al. 1984). P. pastoris has been genetically modified and is used to produce antimicrobial peptides (AMP) (Shrivastava et al. 2023). This synthetic protein has been successfully isolated and proved useful against E. coli-based infections (Cao et al. 2018).

Fungi

Yeast has certain limitations as they tend to hyperglycosylated secreted proteins, reducing its half-life in vivo, and affecting its efficacy. Also, the expression levels and plasmid stability were found to be low, limiting their use, for example- in S. cerevisiae. Therefore, filamentous fungi with a low degree of hypermannoglycosylation can be used during glycosylation. High levels of post-translationally modified therapeutic proteins can be achieved using filamentous fungi such as Aspergillus niger (Ward et al. 2004). Trichoderma reesei is another important host for the synthesis of enzymes such as cellulases and hemicellulases with high yield and production efficacy. Engineered protease deletion strains have been used to synthesize several therapeutic proteins such as bovine chymosin, Ab Fab fragments, interferon α2b and IgG Abs (Belén et al. 2020). Protein expression platforms based on the glyceraldehyde-3-phosphate promoter have been developed in A. unguis. High levels of secreted protein were observed using heterologous signal peptides. This expression system was used for the production of Human interferon β (Madhavan et al. 2017). Matthews et al. (2017) also report the production of human lactoferrin, mucor rennin, and proteinases in A. oryzae (Matthews et al. 2017). Alkaline protease and hirudin can be effectively synthesized by Acremonium chrysogenum (Zhang and Lan 2018). High amounts of therapeutic proteins have been produced using Chrysosporium which makes very little protease (Legastelois et al. 2017).

The recombinant hepatitis B vaccine produced in the non-conventional yeast H. polymorpha has been commercialized (Seo et al. 2008). The H. polymorpha-produced hepatitis B virus antigen (HbsAg) was found to be assembled into yeast-derived lipid membranes. Previous studies have indicated that this lipoprotein particle structure is essential for the antigenicity of the HBsAg (Rutgers et al. 1988). The methanol induction condition, which is routinely used for high-level production of recombinant proteins in the methylotrophic yeast H. polymorpha, is favorable condition for lipid membrane formation and thus advantageous to produce the recombinant hepatitis B vaccine with more desirable antigenicity. Another methylotrophic yeast P. pastoris was recently approved as a host for biopharmaceutical production. The first recombinant biopharmaceutical, a Kallikrein inhibitor (Kalbitor), developed in P. pastoris was approved by the FDA in 2009, and several others are undergoing evaluation in clinical trials (Walsh 2010). Table 3 summarizes the examples of various microbes, such as bacteria, yeasts, and fungi, with therapeutic proteins and their clinical uses.

Table 3.

Summary of some microbial therapeutic proteins and their clinical uses

Microbes Examples Therapeutic Protein Clinical Use References
Bacteria E. coli Interleukin-1 receptor antagonist Auto-immunity disorder Kamionka (2011), Adan Gökbulut and Arslanoğlu (2013), Ahmad et al. (2014a), Wang et al. (2014)
L-asparaginase Lymphoma, mast cell tumor
Insulin Diabetes mellitus
Streptokinase Acute myocardial infarction
Human growth factor Hypopituitary dwarfism
INF-α Leukaemia, hepatitis-B, cancers
INF-β Sclerosis
INF-γ Chronic glaucomatous disease
IL-2 Renal cell carcinoma
Erwinia sp., L-asparaginase Lymphoma, mast cell tumor Pieters et al. (2011)
Pseudomonas sp., P. putida Methionine gamma- lyase, L-glutaminase Infectious disease, cancer, Leukemia Sato and Nozaki (2009)
Bacillus sp., L-glutaminase Leukemia Potla Durthi et al. (2019, Gomaa (2022)
Aeromonas veronii L-glutaminase Lymphocytic leukemia Jesuraj et al. (2017)
Bacteroides thetaiotaomicron α-galactosidase Immune response modification Chaudet et al. (2012)
Yeast Saccharomyces cerevisiae Hepatitis B vaccines H. influenzae type B and hepatitis B infection in infants Martemyanov et al. (2001), Ahmed et al. (2013)
Lepirudin Heparin-induced thrombocytopenia type II Kim et al. (2014)
Desirudin Venous thrombosis Eriksson et al. (1997)
Insulin Diabetes mellitus Adan Gökbulut and Arslanoğlu (2013)
HPV vaccine Cervical cancer caused by human papillomavirus (HPV) Wang et al. (2020)
Rasburicase Hyperuricemia Cammalleri and Malaguarnera (2007)
PDGF Lower extremity diabetic neuropathic ulcer Kim et al. (2014)
HGH Dwarfism, pituitary turner syndrome Kim et al. (2014)
GM-CSF Cancer, bone marrow transplant Kim et al. (2014)
Glucagon Hypoglycaemia Kim et al. (2014)
GLP-1 Type-2 diabetes Kim et al. (2014)
Insulin aspart and Insulin detemir Diabetes mellitus Furman (2017)
Pichia pastoris Ecallantide Hereditary angioedema Craig et al. (2015)
Insulin Type 2 diabetes Polez et al. (2016)
Human serum albumin Blood volume expansion Zhu et al. (2021)
Hepatitis vaccine Hepatitis B Rahimi et al. (2019)
INF-α 2B Hepatitis C and cancer Katla et al. (2019)
Ocriplasmin Vitreomacular adhesion (VMA) Baghban et al. (2020)
Anti-IL- 6R Ab Rheumatoid arthritis Van Roy et al. (2015)
Anti-RSV Ab Respiratory syncytial virus (RSV) infection Kim et al. (2014)
HB-EGF Treatment of interstitial cystitis/bladder pain syndrome (IC/BPS) Kim et al. (2014)
Collagen Medical research reagent/ dermal filler Xiang et al. (2022)
Hansenula polymorpha HBV vaccine Hepatitis B Caetano et al. (2017)
Yarrowia lipolytica Pancrelipase Exocrine pancreatic insufficiency Turki et al. (2010)
Fungi Lentinula edodes α-galactosidase Treatment of fabry disease, blood group conversion and removal of α- type immunogenic epitopes in xenotransplantation Xu et al. (2019), Bhatia et al. (2020)
Schizophyllum commune Hydrophobin, Potential for modification of hydrophobic nanomaterials and solubilization of lipophilic drugs Bayry et al. (2012)
Trametes sp. Laccase Natamycin functionalization Tong et al. (2007), Ji et al. (2022)
Taxomyces andrenae, F. oxysporum and A. niger Paclitaxel (Taxol) Cancer Jozala et al. (2016)
A. foetidus β- galactosidase Lactose intolerance, GM1-gangliosidosis Weesner et al. (2022)
Monascus ruber, A.terreus Lovastatin (statin) High blood cholesterol and reduce the risk of cardiovascular disease Aravindan et al. (2008)
M. hiemalis, A. niger, A. flavus,A. nidulans, A. terreus L-asparaginase Acute lymphoblastic leukemia and non-Hodgkin lymphoma El-Gendy et al. (2021)
Claviceps purpurea Ergot alkaloids Parkinson’s disease, cluster headaches and migraine Meade et al. (2022)
Hericium erinaceus Hericenones and erinacines (cyathane derivatives) Alzheimer’s and Parkinson’s disease Meade et al. (2022)
Aspergillus and Penicillium species Proteases/proteinases/peptidases Cardiovascular disease, emerging agents in the treatment of sepsis, digestive disorders, inflammation, cystic fibrosis, retinal disorders, psoriasis Shankar et al. (2021)
Penicillium nalgiovense Amphotericin B (AMP B) Antifungal, treatment of invasive fungal infections mucormycosis, aspergillosis, blastomycosis, candidiasis, coccidioidomycosis, and cryptococcosis Svahn et al. (2015)
P. griseofulvum Griseofulvin Antifungal, treatment of dermatophytoses Aris et al. (2022)

Microalgal source

Microalgae represent a promising platform for developing recombinant proteins with therapeutic value due to their safety and capacity to produce extracellular vesicles (Banerjee and Ward 2022). Microalgae can be cultured in enclosed systems. Microalgae, like plants, have solid cell walls that protect them from severe gastrointestinal conditions. Microalgae have built-in benefits such as a fast growth rate, low and inexpensive input requirements such as light source, water source, carbon source, supplementary nutrients, and photoautotrophic nature. Transformation and production of recombinant therapeutic proteins in microalgae require minimal time (Kaye et al. 2015; Hempel et al. 2017). Microalgae can produce a variety of bioactive compounds, such as carotenoids, polysaccharides, vitamins, and lipids. In addition, they have a wide range of potential applications in the biomedical, pharmaceutical, cosmetics, environmental, and animal feeding industry (Rizwan et al. 2018; Fu et al. 2019). There are three transformable sites in microalgae, i.e., chloroplast, nucleus, mitochondria, and out of these, nucleus and chloroplast sites are nowadays attracting the attention of scientific community for the development of recombinant proteins (Dehghani et al. 2020). Transgene expression in the model microalga via chloroplasts Chlamydomonas reinhardtii can produce protein concentrations > 5% of total soluble protein higher than plant systems (Manuell et al. 2007; Specht and Mayfield 2014; Barbosa et al. 2023). Due to their safety and solitary chloroplast, which expresses proteins with great accumulation, microalgae (particularly C. reinhardtii) are appropriate options to be employed as vaccine carriers (Khavari et al. 2021). In a related study, Demurtas et al. (2013) engineered C. reinhardtii chloroplast to express the E7GGG protein needed for therapeutic vaccinations via inserting the HPV16 E7 protein gene into the (C. reinhardtii) chloroplast's genome (Demurtas et al. 2013). Nowadays, the majority of monoclonal antibodies produced by cell lines derived from Chinese hamster ovary are pretty expensive and carry a risk of pathogen contamination. Therefore, microalgae are regarded as effective alternative host cells because of their advantages (Khavari et al. 2021).

These microalgae can appropriately handle the post-translational changes of human recombinant proteins than bacterial cells. In a study, Hempel et al. (2017) developed a monoclonal IgG antibody against the Marburg virus's nucleoprotein, which is a significant cause of hemorrhagic fever in western Africa (Hempel et al. 2017). Microalgal antiviral substances have an impact on viral infections at various stages. For instance, the first step is hampered by sulfated polysaccharides (Fu et al. 2016). In a study on this subject, Hayashi et al. (2019) were able to extract a monogalactosyl diacylglycerol (MGDG) from Coccomyxa sp. that led to physical alterations in HPV that caused it to encapsulate. The virus was unable to connect to the host cell, showing that MGDG has antiviral properties (Hayashi et al. 2019). A sulfated polysaccharide called fucoidan, which is extracted from various microalgae including Fucus vesiculosus, Sargassum henslowianam, Cladosiphon fucoidan, and Coccophora longsdorfii, inhibits angiogenesis and metastasis by lowering down kinase activity and activating caspase-3/7 in human colon cancer, melanoma, breast cancer, etc. (Deniz et al. 2017). Miceli et al. (2019) reported selective apoptosis of colon cancer cell line (HCT-116) and haematological cancer cell line (U-937) using monoacylglycerides extracted from Skeletonema marinoi through the activation of caspase-3/7. Still, no apoptosis was induced in normal cells (Miceli et al. 2019). Astaxanthin from microalgae, Haematococcus pluvialis can reduce the cytokine storm, averting acute respiratory distress syndrome (ARDS) and acute lung injury (ALI) in COVID-19 patients (Talukdar et al. 2020).

For microalgal production of recombinant therapeutic proteins, chloroplast transformation is a significantly utilized approach primarily for some of the inbuilt advantages such as the single promoter for expression of multiple genes, no gene silencing, no position-effect change, elevated stages of transgene expressions, etc. (Sproles et al. 2021). However, the major issue with the microalgal production of therapeutic proteins is low yield. Various genetics-based tools and approaches were made available to improve protein expression. Banerjee and Ward have summarized different ways and techniques for other microalgal species to enhance the yield of therapeutic proteins (Banerjee and Ward 2022). Agrobacterium-mediated transformation also proved to be a reliable method for transgenes to be transferred to the genome of many microalgal species. Using AMT process, active human interleukin-2 (hIL-2) was produced using C. reinhardtii, C. vulgaris, and D. Salina (Dehghani et al. 2020). A significant challenge for transgenic strain production of microalgae is randomized insertion of transgene leading towards heterogenicity. High-throughput techniques will be essential and needed to speed up the screening process and choose the finest clones. Microalgae are currently being grown in bioreactors for the expression of recombinant proteins. However, the consequences of the cultural conditions are appallingly understudied (Banerjee and Ward 2022). The scaling up of these cultures in bioreactors in a manner compliant with good manufacturing practices is one issue with the use of obligate phototrophic species of microalgae for the manufacture of recombinant proteins (Banerjee and Ward 2022). Table 4 summarizes some microalgal therapeutic proteins and their clinical applications.

Table 4.

Summary of some microalgal therapeutic proteins and their clinical uses

Sr. no Host (microalgae) Therapeutic proteins Clinical use References
1 P. tricornutum CL4mAb with ER retention signal against hepatitis Hepatitis B virus Hempel et al. (2011), Vanier et al. (2015)
2 P. tricornutum CL4mAb without ER retention signal against hepatitis Hepatitis B virus Hempel et al. (2011), Vanier et al. (2015)
3 P. tricornutum IGA against Marburg virus Marburg virus Disease Hempel et al. (2017)
4 Chlorella sp. Fish growth hormone Kim et al. (2014)
5 Chlorella sp. Trypsin-Modulating Oostatic factor Mosquitoes decapeptide Borovsky et al. (2016)
6 C. reinhardtii Human erythropoietin Anemia Eichler-Stahlberg et al. (2009)
7 P. tricornutum Hepatitis B surface Ag Hepatitis B Hempel et al. (2011)
8 Chlamydomonas sp. HIV-1 P24 Ag Barahimipour et al. (2016)
9 Chlorella sp. IBDV VP2 protein Reddy et al. (2017)
10 C. reinhardtii IGA against herpes simplex virus Herpes simplex virus Mayfield et al. (2003)
11 C. reinhardtii IgG1 against Anthrax Anthrax Tran et al. (2009)
12 C. reinhardtii Immunotoxin against Lymphoma Lymphoma Tran et al. (2013)
13 C. reinhardtii Human growth hormone Turner syndrome Wannathong et al. (2016)
14 C. reinhardtii Angiotensin II fused to Hepatitis B virus capsid antigen (HbcAg) Hepatitis B Soria-Guerra et al. (2014)
15 C. reinhardtii, C. vulgaris, and D. salina Human interleukin 2 Cancer Dehghani et al. (2020)
16 P. tricornutum RBD (Receptor binding domain) of SARS-CoV-2 COVID-19 Slattery et al. (2022)
17 C. reinhardtii Human interferon alpha 2a Cancer Commault et al. (2020)
18 C. reinhardtii RBD SARS-CoV-2 COVID-19 Berndt et al. (2021)
19 C. reinhardtii Spike glycoprotein of SARS-CoV-2 COVID-19 Kiefer et al. (2022)
20 Schizochytrium sp. BCB (multi-epitope protein) Breast cancer Hernández-Ramírez et al. (2020)
21 C. reinhardtii Human VEGF-165 (human vascular endothelial growth factor) Wound healing Jarquín-Cordero et al. (2020)

Expression hosts for the production of therapeutic proteins

Recombinant therapeutic proteins represent a major and significant percentage of approved drugs. However, the effectiveness of these drugs may be compromised due to their short in vivo half-lives triggered by proteolysis and abrupt disposal. To get the desired therapeutic effect, large doses must be administered regularly, which entails a higher risk of toxicity, considerable patient discomfort and suffering, and a high cost (Du et al. 2019). PEGylation (Bailon and Won 2009) and the insertion of fusion tags such as Fh8, SUMO, and immunoglobulin (Ig) G Fc fragments (Levin et al. 2015) have been developed and used in the past to boost therapeutic protein in vivo activity and half-life (Kontermann 2011; Du et al. 2019). PEGylation of therapeutic proteins is a prevalent approach in the pharmaceutical business. However, studies suggest that the immunogenicity of PEG and the formation of anti-PEG antibodies, as well as the buildup of unused PEG in body tissues, are some of the harmful and devastating consequences (Verhoef et al. 2014; Chen et al. 2021). Therefore, there is a requirement of other protein modification techniques. PTM glycosylation (either N- or O-linked glycan) is naturally present in eukaryotic cells. These glycans improve protein stability, solubility, half-life, and biological functioning, among other things (Varki 2017). However, glycoengineering-based modifications and investments are required to improve therapeutic protein efficacy (Solá and Griebenow 2010). However, different expression methods can produce glycoproteins with a wide range of glycoform compositions. Some of the expression systems, viz., bacteria, yeast, insects, mammals, transgenic animals, and transgenic plants, are briefly discussed for therapeutic protein production.

Bacterial expression systems

Bacterial expression systems are one of the most critical industrial expression hosts due to their advantages of quick growth, well-characterized genetics, a well-established safety background, and high protein production. However, the restricted environment of bacterial cells, as well as the lack of PTM, are issues associated with bacterial expression hosts (Du et al. 2019). Nevertheless, cellular engineering approaches have enabled the expression of human proteins in bacterial cells. For example, the commercial strain of E. coli Shuffle from New England Biolabs offers an oxidizing cytoplasmic environment and promotes proper protein folding by increasing disulfide bond formation (Lobstein et al. 2012). Co-expression of E. coli with Saccharomyces cerevisiae's chaperone sulfhydryl oxidase and human protein disulfide isomerase (hPDI), as reported by (Nguyen et al. 2011), also improves protein folding. Attempts have been made to overcome the fact that E. coli does not natively manufacture N- or O-glycosylated proteins with mammalian-style glycans by demonstrating the feasibility of N- and O-linked glycosylation in E. coli (Pandhal and Wright 2010; Merritt et al. 2013). For instance, the expression of several glycosyltransferases in E. coli allowed for the synthesis and transfer of a tri-mannosyl chitobiose N-glycan to the designed target eukaryotic protein (Valderrama-Rincon et al. 2012).

Yeast expression systems

Yeast, as a unicellular eukaryotic microbial host cell, provides unique advantages for the production of biopharmaceutical proteins. The use of yeasts enables an ideal combination of steady development of the primary media in large-scale bioreactors, post-translational modifications, and ease of genetic manipulation (Mattanovich et al. 2012). Since the initial industrial manufacture of recombinant human insulin in S. cerevisiae in 1987, several pharmaceutical medicines have been marketed, many of which are highly successful (Walsh 2010). Non-saccharomyces yeast species are effective alternatives to S. cerevisiae as hosts for biotechnological applications owing to their superior secretion capability and faster growth on inexpensive carbon sources. Non-conventional yeasts have used conventional methods for developing toolboxes for genetically engineering host strains to increase protein production capacity, boost product quality, and simplify downstream processing (Mattanovich et al. 2012; Krainer et al. 2012; Saraya et al. 2014).

S. cerevisiae expression systems yield about 15% of protein-based biopharmaceuticals for human application (Wang et al. 2017). S. cerevisiae is superior to bacteria, other yeasts, and filamentous fungi in key physiological features related to the production of commercial ethanol. This physiological flexibility includes resistance to high osmotic pressure, high ethanol, and sugar concentrations, and a broad pH tolerance (Tesfaw and Assefa 2014; Baghban et al. 2019). Among eukaryotic hosts, S. cerevisiae is the most popular for investigating gene expression regulation, secretory pathways, and other associated issues (Baghban et al. 2019). Whenever heterologous glycoproteins are synthesized in S. cerevisiae, the resulting glycan structure is often hyper-mannosylated, which can reduce activity and increase immunogenicity. The synthesis of recombinant cellulases is improved and boosted when the Mnn2p and Mnn11p genes in the glycosylation modification pathway are disrupted, as revealed by Tang et al. (2016) (Tang et al. 2016). S. cerevisiae is a yeast cell that is utilized in the production of hepatitis B and human papillomavirus vaccines, both of which produce a protective immune response against wild-type viruses (Bill 2015). In S. cerevisiae, expressed proteins are frequently N and O-hyperglycosylated, which may impact protein immunogenicity (Rasala and Mayfield 2015). To address the issue of protein expression, methylotrophic yeasts such as Hansenulla polymorpha and Pichia pastoris (P. pastoris; syn. Komagataella phaffii) have been produced in recent years. P. pastoris has been the most popular because of its low cost and expression host system. This microorganism can create large amounts of recombinant proteins with glycosylation, which is similar to that of mammalian cells (Balamurugan et al. 2007). Overall, the advantages of protein production by the P. pastoris system include proper folding (in the endoplasmic reticulum, ER) and secretion of recombinant proteins to the cell's external environment (via Kex2 as signal peptidase) (Yang et al. 2013). Because some proteins produced by their original host are secreted out of the cell, P. pastoris is excellent for the creation of recombinant proteins because it has a secretion system (Ahmad et al. 2014b).

Insect cells as expression host

Insect cells have the benefit of executing PTMs that are more comparable to those in humans and are able to produce recombinant proteins with reasonable cost and yield (Dana 2018). Insect cells can be infected with baculoviruses, which are double-stranded circular DNA viruses with arthropods as host. Baculovirus-mediated gene expression in insects is a method of choice and is cost-effective, giving a much higher yield of recombinant protein compared to other systems. It is possible to produce large proteins, resulting in the production of correctly processed and biologically active proteins. Baculovirus-based vectors used in the insect cell expression system are primarily able to integrate the transgene into the insect cells' genomes (Osz-Papai et al. 2015; Kost and Kemp 2016). However, there are some problems associated with insect hosts, such as the creation of baculovirus vectors, which could take a long time and be difficult to create (Osz-Papai et al. 2015; Dana 2018). Humans do not possess the paucimannosidic glycans that are added by insect cells (Burnett and Burnett 2020). Insect cells also have limitations in performing post-translational modifications as it performs non-syalated N-linked glycosylation. All the other optimizations need to be perfect as yield depends upon the virus titer and the time taken from infection to expression. Insect cells are preferred when active protein is difficult to obtain in E. coli system. Glycosylation is another problem which is encountered when insect cells are used for the production of recombinant human glycosylated proteins (Gupta and Shukla 2016). Extensive genetic engineering is required to produce human-like glycosylation in insect cells (Clausen et al. 2022).

Mammalian cell expression system

Among all approved recombinant protein-based biopharmaceuticals, mammalian cells dominate the other recombinant protein-expression systems (Owczarek et al. 2019). Mammalian cells can express large and complex recombinant proteins. Mammalian cells have the advantage of performing PTMs correctly, and they secrete recombinant protein in their natural form into the medium (Gupta and Shukla 2016; Dana 2018). Although the glycosylation patterns of Chinese hamster ovary cells are generally regarded as safe (i.e., they don't include the hyper-immunogenic α-gal epitope), they do have limitations. For instance, they are inadequate production hosts for potent anti-inflammatory antibodies because they lack 2,6-sialyltransferase activity. The fact that CHO cells produce the Neu5Gc form of sialic acid despite being relatively moderately immunogenic raises concern (Ghaderi et al. 2010, 2012).

The majority (~ 84%) of the approved biopharmaceutical products on the market today are produced in CHO cell-based systems (57 products), with the remaining antibodies being expressed in NS0 cells (9 products) or Sp2/0 cells (2 products) (Walsh 2018). In mammalian cell lines, PTM is present, although their glycosylation pattern differs from the human type. Protein expression is also being investigated in the human cell lines- HEK293, HKB11, PER.C6, HeLa, and CAP (Bandaranayake and Almo 2014; Dyson 2016; Dumont et al. 2016; Hu et al. 2018; Gupta et al. 2019; Hunter et al. 2019). The expression of proteins modified in a way similar to humans is stimulated in a human cell line. HeLa cells were used to generate a recombinant protein effectively encoding the fully glycosylated form of connective tissue growth factor CCN2 (Nishida et al. 2017). However, large-scale commercial production of these cells is still in the research and development stages. Contamination with animal viruses is another issue with this method. Culture medium formulation for a cell line is challenging due to the need for several components, such as growth hormones, amino acids, reducing agents, and vitamins. There are several subtypes of CHO cells, including CHO-K1, CHO-S, CHO-DG44, and CHO-DXB11.

Reinhart et al. (2019) examined CHO-K1, CHO-S, and CHO-DG44 host cell-specific differences in mAb expression in batch, fed-batch, and semi-continuous perfusion cultures, revealing CHO cell line-specific preferences for mAb production. The host cell line and media also affected the quality attributes of mAb. It has also been demonstrated that cell engineering prevents the accumulation of ammonium and lactate and enhances cell proliferation (Kim and Lee 2007a). The cell line is optimized through codon optimization and other methods (Zhu 2012). In addition, glycoengineering is utilized to produce the desired glycoform of a protein to enhance its efficacy and create a high-quality product (Lalonde and Durocher 2017; Wang et al. 2017; Heffner et al. 2018). For mammalian expression systems, gene-editing technologies such as CRISPR/Cas9 have been effectively implemented to improve product quality. Li et al. (2019) conducted a study in which C1s protease was inactivated using CRISPR/Cas9 for the production of recombinant HIV envelope protein gp120 in CHO cells. Yang et al. (2019) identified CRISPR/Cas9-mediated site-specific integration as an efficient and dependable method for establishing stable recombinant HEK293 cell lines for biopharmaceutical production. Gene modification with CRISPR/Cas9 allowed HEK293 cells to bioprocess media without antibiotics (Román et al. 2019). CRISPR/Cas9 was used to generate Anxa2- and Ctsd-knockout CHO cell lines, which led to the elimination of the corresponding host cell protein (HCP) in cell lysates without compromising cell growth or viability, paving the way for the successful creation of recombinant proteins (Fukuda et al. 2019). CRISPR/Cas9-mediated deletion of microRNA-744 enhanced antibody titer in CHO production cell lines (Raab et al. 2019). Among gene-editing tools, CRISPR/Cas9 and recombinase-mediated cassette exchange (RMCE) technology will contribute most to the advancement of glycoprotein production in the near future.

Transgenic organisms as expression host

Transgenic animals

Transgenic animals have been used to express recombinant protein-based medicines, such as monoclonal antibodies (mAbs), vaccinations, hormones, enzymes, and growth factors. Animals with a transgene incorporated into their DNA that codes for a recombinant protein can pass that protein on to their progeny. Milk from transgenic mammals and eggs from transgenic chickens are two examples of where proteins are sourced nowadays (Moura et al. 2011; Maksimenko et al. 2013; Owczarek et al. 2019). The natural secretion of recombinant proteins provides correct post-translational modifications (PTMs) in this method. However, creating transgenic animals raises serious moral concerns. Protein preparations from transgenic animals may contain zoonotic diseases (Wang et al. 2013; Bertolini et al. 2016). Transgenic cattle have the potential to be used as bioreactors for the industrial production of protein in milk. However, there are currently inefficiencies in the methods used to achieve this goal (Monzani et al. 2016). To create animals that generate recombinant proteins in milk, (Shepelev et al. 2018) investigated targeted genome-editing tools and other methods for developing transgenic animals.

Transgenic plants

Recombinant biopharmaceutical production can be improved with the use of transgenic plants. Low cost, safety (low risk of contamination with animal pathogens), easy scale-up, stability, presence of metabolites, and ability to produce N-glycosylated proteins are some of the benefits of this system (Fahad et al. 2015; Yao et al. 2015; Xu et al. 2016; Lomonossoff and D’Aoust 2016; Park and Wi 2016; Buyel et al. 2017). In recent years, cancer immunotherapy drugs derived from plants have been developed (Chen et al. 2016; Hefferon 2017). The production and quality of plant-produced biopharmaceuticals can be improved by paying attention to a few key parameters, such as host plants, expression cassettes, subcellular localization, post-translational modifications, and protein extraction and purification strategies. Glycoengineering has been studied extensively to lessen the immunogenicity of plant-made recombinant proteins (Moustafa et al. 2016). Contamination of the final product by harmful plant metabolites and pesticides is a significant drawback of this approach. Controlling the level of transgene expression and the complexity of the purification process are additional challenges associated with this system. Most recombinant proteins are produced in plant systems, including cell culture, tissue-based systems, and transgenic plant building. Bacterial infection (agro-infection) or viral infection, as well as direct techniques like biolistic bombardment and the PEG-mediated procedure, are commonly used to introduce the transgene into plant cells. The ability to express the recombinant protein in the desired cell compartment or plant organ is a significant benefit of these expression systems. The glycosylation pattern of human therapeutic proteins generated in plants is generally more similar to plants than to humans. This problem is being tackled with the help of glycoengineering (Fischer et al. 2018; Owczarek et al. 2019). Both traditional and cutting-edge methods for improving the production of recombinant proteins, such as the use of gene-editing tools coupled with the insertion of target genes in euchromatin genome areas, can be used (Rozov and Deineko 2019).

Edible vaccines have been derived from transgenic plants such as rice, bananas, peas, potatoes, lettuce, and corn. Transgenic rice cell culture has used genetic engineering to produce proteins at levels as high as 247 mg/l (α1-antitrypsin) and 100 mg/l (antibodies) (Loh et al. 2017; Owczarek et al. 2019). Human recombinant β-glucocerebrosidase (taliglucerase α-approved by FDA in 2012) enzyme was produced on a large scale in carrot (Daucus carota) cell culture (ProCellEx™) for the treatment of Gaucher disease (Tekoah et al. 2015; Moustafa et al. 2016). The world's first plant-derived IgA mAb that detects the surface antigen I/II of the bacteria Streptococcus mutans has been licensed in Europe and is used to prevent tooth decay (Larrick et al. 2001; Loh et al. 2017). Biopharmaceuticals derived from plants are in various phases of clinical trials or implementation on the market (Yao et al. 2015; Park and Wi 2016; Dirisala et al. 2017; Owczarek et al. 2019). Examples include HAI-05 [Influenza Vaccine- for Influenza A virus H5N1, tobacco (Nicotiana tabacum) as the host plant, phase II], Insulin (SBS-1000) for diabetes, safflower (Carthamus tinctorius) as the host plant, phase III], ZMApp (monoclonal antibody cocktail) for Ebola virus, tobacco (N. benthamiana) as the host plant, phase II, human growth hormone for deficiency treatments, host plant-barley seed (Hordeum vulgare), status, commercialization] (Owczarek et al. 2019). The development of a new plant system based on carnivorous plants demonstrated the ability of biomimetic approaches to contribute to the production of novel recombinant proteins. Nevertheless, the modest protein yields disqualified these plants from an industrial platform (Miguel et al. 2019). The establishment of a new plant system based on carnivorous plants demonstrated the ability of biomimetic approaches to contribute to the production of novel recombinant proteins. Nevertheless, the modest protein yields disqualified these plants for an industrial platform (Miguel et al. 2019). Donini and Marusic (2019) (Donini and Marusic 2019) described recent developments in the production of mAbs using plant-based systems such as transgenic plants, tissue and cell cultures, and transient expression systems. Holtz et al. (2015) described a commercial biotherapeutics manufacturing facility for plant-based pharmaceuticals. Various methods for the plant-based production of recombinant proteins and recent advances in the development of plant-based therapeutics and biologics for the prevention and treatment of human diseases have also been described (Loh et al. 2017; Rozov et al. 2018; Schillberg et al. 2019; Tripathi and Shrivastava 2019).

Production of therapeutic proteins and process optimization

We are all aware of the global health crisis caused by infectious illnesses. Therefore, there is an urgent need for the mass production of therapeutic proteins to combat these epidemics. Recombinant DNA technology has allowed the production of therapeutic proteins in heterologous expression systems (mammalian cells, bacteria, yeast, transgenic plants, and animals at laboratory and large scale) that can be used as therapeutics, vaccines, and diagnostic reagents. There are several advantages of using these recombinant proteins, such as low immunological rejection, low risk of infection, and reduced exposure to animal or human diseases (Leader et al. 2008). For the production of these recombinant proteins, some recent advances have also been made in the bioprocessing sector, such as the use of high-throughput devices (microtiter plates, bubble column or microplate-based mini-bioreactors, stirred mini-tank bioreactors), continuous upstream processing, quality by design, continuous chromatography to achieve a good quality product with higher yield (Tripathi and Shrivastava 2019). The strategies for microbial production of proteins are of great importance in developing application-based research and explaining the novel purification strategies used for industrial production. These strategies or processes play a vital role in effective therapeutic protein development at the industrial level. Some of the strategies adopted for the optimum production of therapeutic proteins have been discussed below:

Bioreactor conditions

Optimization of therapeutic proteins is primarily based on fed-batch cultures after initial development via a batch process, which provides enormous insight into the decoupling growth and production phase, improved cellular activity control, and balanced media uptake (Vandermies and Fickers 2019). Factors such as starvation, overfeeding, and high concentration and accumulation of media and nutrients for cells must be considered throughout production to maintain the constant growth of products. Trassaert et al. (2017) observed that reactor-scale continuous processes can be utilized as exploratory chemostats to determine the optimal promoter induction settings. At the industrial level, however, issues with continuous processes, such as contamination and product instability, must be addressed. By transitioning from shake-flask to bioreactor cultures, the oxygenation conditions and medium uniformity are enhanced (Gasmi et al. 2012). This transition from shake-flask to bioreactor increases productivity by 3 to 5 folds, and even more for complex non-enzymatic therapeutic proteins, such as a 416-fold increase in human interferon concentration after scaling up the culture from 250 ml to 5L batch reactor (Gasmi et al. 2012).

Complex media offers increased biomass and protein production yields. Still, non-defined composition and batch-to-batch variability affect production, which is why defined media are utilized for controlled processes such as the production of therapeutic proteins (Gasmi et al. 2012; Vandermies and Fickers 2019). The availability of oxygen is another crucial factor affecting production processes. The concentration of dissolved oxygen (DO) in culture media influences cell physiology. It provides information about the growth phase (exponential or stationary) and the health and condition of the cell population. While oxygen needs to diminish at the end of culture owing to decreased metabolic activity, it increases during the exponential growth phase, paralleling the expansion of substrate metabolism by a growing number of cells. In addition, abnormally elevated DO levels indicate cell starvation in media (Vandermies and Fickers 2019). Fixed agitation and aeration levels are the most uncomplicated operational designs for processes that do not require a large amount of oxygen (Darvishi et al. 2017). Manually or automatically, air flow and, more frequently, agitation are regulated for optimal oxygen transfer to the culture fluid (as measured by the oxygen transfer rate, OTR). Inadequate pH levels may affect the production of therapeutic proteins and be detrimental to the activity and stability of the proteins (Celińska et al. 2015, 2016). Although biomass load in the bioreactor receives less attention, this parameter influences the production process by affecting the growth kinetics of microorganisms. In fed-batch systems, adequate nutrient uptake results in an increase in cell density (Celińska et al. 2017; YaPing et al. 2017).

Cell line development and clone selection

During cell-line development, several factors were taken into consideration, such as the selection of host cells, expression vectors, transfection, and selection methods. High throughput devices such as ClonePix (Thermo) and FACS (BD and Beckman) are now being used for cell line development and screening. The selection of the expression system is based on its ability to ensure and meet high productivity along with quality criteria. Among the various expression systems in industries, E. coli (bacteria), S. cerevisiae, and P. pastoris are mainly used to produce therapeutic proteins and are in commercial use worldwide. The most preferable and widely used host for the production of therapeutic proteins is E. coli due to its low cost, rapid growth, good productivity, and well-known biochemistry (Baeshen et al. 2014; Gupta and Shukla 2017a). But there are some limitations also, such as lack of proper post-translational modifications (PTMs), formation of inclusion bodies (IB), production of endotoxins, etc. These limitations can be avoided by adding fusion tags such as Fh8, SUMO, TRX, His at N- or C- terminal, co-factors, co-expression of the protein with molecular/chemical chaperons, which enhances the solubility of proteins and help in affinity purification (Paraskevopoulou and Falcone 2018).

Further, lowering the temperature and glycoengineering E. coli cells also leads to better protein solubility and increases protein folding. However, some PTMs have been done in E. coli to produce therapeutic proteins. The addition of diverse carbon and nitrogen sources along with acetate metabolism knockout strains can redirect E. coli carbon-fluxes to different pathways and result in a fivefold increase in protein production (Chung et al. 2017; Huang et al. 2019; Klein et al. 2019; Lozano Terol et al. 2019).

Another expression system, i.e., S. cerevisiae can be rapidly grown in protein-free media, presence of PTMs, ability to secrete extracellular product. The gene encoding mannosyl transferase has been knocked out and ALG3/ALG11 double knockout was generated to prevent hypermannosylation of expressed protein (Parsaie Nasab et al. 2013). Then, the GlycoSwitch platform was developed in which hypermannosylation gene of yeast is removed and used to produce glycosylated proteins. However, the lower yield limits its commercial use. Pichia pastoris (Komagataella phaffi or K. pastoris) is methylotrophic yeast which as the ability to produce properly folded and functional proteins. They also provide reduced protein glycosylation and achieve high cell densities (Looser et al. 2015; Yang and Zhang 2018; Juturu and Wu 2018; Werten et al. 2019). However, proteolytic degradation or truncation of the product leading to low yield and loss of biological activity are the major problems associated with P. pastoris, which can be reduced using yeast peptone, casamino acids, protease inhibitors, lowering of pH and temperature is done. Nowadays, mammalian cell line such as CHO (Chinese hamster ovary cells) cell line dominate the other protein expression systems as they can express large and complex recombinant proteins. CHO cells having different lineages- CHO-K1, CHO-S, CHO-DG44 and CHO-DXB11 are mostly used as a host to produce recombinant therapeutic proteins including monoclonal antibodies (mAbs) and fusion proteins (Kelley 2009; Rita Costa et al. 2010; De Jesus and Wurm 2011; Fan et al. 2013; Dumont et al. 2016; Gupta and Shukla 2016; Zhu et al. 2017; Sharker and Rahman 2021). About 70–80% of all recombinant therapeutic proteins have been produced in CHO cells (De Jesus and Wurm 2011; Gupta and Shukla 2016).

Other factors such as cell growth pattern, stable production, cultivation in serum-free medium as a suspension culture, scalability in bioreactor, attributes of quality of product are also considered. The glycosylation pattern in mammalian cell lines is different from human-type glycosylation. The expression of proteins with human-like PTMs is enhanced by a human-cell line. Cell engineering helps to avoid accumulation of ammonium lactate and improves cell growth (Kim and Lee 2007b) and the cell line is optimized by codon optimization. A new modular cloning system has been developed for flexible de novo plasmid assembly, MoCloFlex which can be used to plan, build, and isolate a custom plasmid within 24 h, leading to low cost and less time consumption (Klein et al. 2019). CRISPR/Cas9 has also been used for the chromosomal integration of large DNA into E. coli successfully and able to integrate functional genes in diverse E. coli strains (Chung et al. 2017). There are some recent developments done in the field of metabolic engineering which includes the use of knock-in (KI) and knock-out (KO) gene-editing tools for successful clone and product development. Using CRISPR/Cas9, C1 protease was inactivated to produce recombinant HIV envelope protein group 120 in CHO cells (Li et al. 2020). CRISPR/Cas9 assisted native end-joining and editing offered a simple strategy for efficient genetic engineering in E. coli (Huang et al. 2019; Amiri et al. 2023). It is now possible to integrate large DNA in E. coli using CRISPR/Cas9 and can also be used for yeast engineering for site specific gene integration or knock-out of certain unwanted genes in an improved protein-production (Utomo et al. 2021; Ingle et al. 2023).

Manufacturing process development

Analysis of the medium spent in the fermentation of microbial system provides an insight into consumption and accumulation of constituents present in the culture supernatant. The nutrients such as carbon sources used in E. coli and yeast culture, can affect cell metabolism, protein production and quality significantly (Seyis and Aksoz 2005). The clone, medium and feed screening plays a crucial role at early stage for high productivity. With the advent of high-throughput cultivation systems such as miniature shaken vessel/ wells/ microtiter plates (MTPs), bubble column or microplate-based mini-bioreactors, and stirred mini-tank bioreactors, it has now become very easy to perform screening assays including optimization of processes which saves time and cost before proceeding to scale up the production of recombinant proteins (Fisher et al. 2019; Tripathi and Shrivastava 2019).

The development process starts with the identification of protein expressing cells, which are first used for small scale and bioreactor culturing to analyze and check the cell growth and protein production levels. Optimization of batch and fed-batch production processes have also been established and improved to a large extent (Schmuck et al. 2021). BioLector (microbioreactor) is being used to perform high-throughput fermentation altogether with system (online) monitoring of the process parameters such as biomass, pH, dissolved oxygen (DO), florescence, etc. It works with non-invasive optical sensors and controls the shaking speed, temperature as well as humidity (Fink et al. 2021). In a study, 32 production clones were characterized in BioLector with production yield of 0–7.4 mg Fab/g of cell dry mass (Fink et al. 2021). In a different study, after comparing 51 cultivations, it was reported that the fully automated HTP cultivation speeds up the identification of the optimal expression systems and process conditions which leads to automated early-stage bioprocess development (Sawatzki et al. 2018) (Fig. 1).

Fig. 1.

Fig. 1

Summary of the process involved in production of therapeutic proteins. The process comprised of cloning and expression, selection of hybrids, single cell cloning, optimization and growth evaluation. Further depending on the nature of protein, supernatant or cell pellets can be collected for extraction process. After purification, new formulations can be developed and packaged

Quality by design (QbD) or design of experiments approach (DOE)

This approach has also been studied and applied for upstream processes in which quality of product is integrated into the manufacturing process. According to one study, QbD resulted in a fivefold increase in target protein titer when compared to the basal medium and demonstrated its efficacy. In this process, a large amount of data was provided in a short span of time (Kumar et al. 2019a). For the process development and analytical evaluation of recombinant proteins, including mAbs, the QbD approach has been used (Pathak et al. 2014; Yu et al. 2014; Narayanan et al. 2019).

To reduce the time and money spent on experiments during upstream process development, the Design of Experiments (DoE) method has been applied to different recombinant protein production process parameters (Papaneophytou and Kontopidis 2012; Hanke and Ottens 2014; Shekhawat et al. 2019). Various design-of-experiments (DoE) methods, including full factorial design, Taguchi orthogonal arrays, fractional factorial design, and the response surface methodology, are used to optimize media to maintain a balance between the different components of the media, thereby increasing protein yield (Shekhawat et al. 2019; Kumar et al. 2019b). In one study, to produce recombinant antibody fragments in the periplasm of E. coli, the signal peptide was chosen using DoE, and the best growing conditions were developed (Kasli et al. 2019). In another study, a specified medium to increase the synthesis of human interferon gamma was developed using RSM (Unni et al. 2019).

Process analytical technology (PAT)

This process is used to ensure that the final product meets the desired specifications by designing, analyzing, and manufacturing control via periodic and/or continuous measurement and check on the critical quality and performance attributes (Shaikh et al. 2018). The reactor design, cell harvesting, process control and analytics are also a part of optimization process and are optimized individually and the main goal is to attain high productivity with defined quality (Gronemeyer et al. 2014; Reyes et al. 2022). To implement PAT for the advancement of continuous cultures, innovation in sensor technology is required, as well as improvements in its configuration and resilience (Fisher et al. 2019).

Downstream processing

Downstream process mainly focusses on purity parameter, i.e., process and product impurity removal as well as yield and productivity (Owczarek et al. 2019). We can use a wide range of techniques, approaches, and technologies for an efficient and cost-effective downstream process, such as building a platform process, QbD and DOE-based high-throughput methods, a single-use system, integrating modelling, and replicating mini-plant/pilot plant facilities. Conventionally, a mAb or any other therapeutic protein is purified using chromatographic techniques (Zydney 2016). The downstream process involves 2 major steps: Filtration and Chromatography. Filtration includes harvest clarification for biomass removal, virus filtration to remove the viruses and tangential flow filtration for protein concentration and polishing step. Primary cell clarification involves centrifugation, TFF-MF (Tangential flow microfiltration), and depth filtrations. TFF-MF is highly efficient process and helps in the removal of whole cell mass and its fragments based on size-exclusion and utilizes microfiltration membrane with pore size of up to 0.65 µm. Single use centrifugation device was developed by KSep, which was fully automated and can recover over 97% of product/ cell biomass, which can be used for harvest clarification of recombinant proteins and vaccines (Gupta and Shukla 2017b). After the harvest clarification, a chromatographic resin is used to capture the expressed protein from upstream bioreactor. There are various chromatographic techniques such as affinity, ion exchange, hydrophobic interactions and size inclusion or gel filtration which are used to achieve a biologically active highly purified product (Wilken and Nikolov 2012).

Affinity chromatography can be used for the purification of tagged proteins, bispecific Abs, cellular products, DNA-based biologics, viral vectors, and viruses using Hexahistidine (His), maltose-binding protein (MBP), glutathione S-transferase (GST) tags (Zhao et al. 2019; Łącki and Riske 2020). Protein A chromatography can be used for mAb purification, but there is certain limitation associated such as leaching of Protein A with non-specific binding of host cell protein (HCP), elevated cost of resins and short lifetime, protein A ligand modification and alternative formats such as monolith membrane and microspheres (Ramos-de-la-Peña et al. 2019). Ion-exchange chromatography can be used as a cost-effective method for purification of recombinant proteins. Impurities such as media components, endotoxins, remaining HCP and DNA, product variants are removed using Cation and anion exchange chromatography (CEX and AEX). The efficacy of a weak anion exchanger was reported on the isolation of rHBsAg VLPs (Virus-like particles) from aggregated structures and found to yield 94–97.5% content of rHBsAg VLPS within the acceptable quality level (Kimia et al. 2019). From yeast crude extract, it resulted in high purity (> 95%). It was also reported that using CEX in overloaded mode, it was possible to remove viruses during the manufacture of therapeutic proteins (Masuda et al. 2019). Hydrophobic interaction (HIC) chromatography depending on hydrophobicity of molecules can be used for purification of recombinant proteins in both small and large-scale purification of hormones and industrial enzymes (Bhuvanesh et al. 2010; Saraswat et al. 2013). In a recent study, the influenza A and B viruses were successfully purified using HIC and gave efficacy of about 96% virus recoveries and about 1.3% residual DNA using 96-well plate method (Weigel et al. 2019). Insulin like growth factor receptor (Levin et al. 2016) from E. coli was purified using Size exclusion/Gel filtration chromatography. In a study, a yellow fever vaccine was prepared using bioreactors, AEX membrane adsorber, a multimodal resin and beta-propiolactone inactivation. The overall recovery rate was 52.7%(Pato et al. 2019).

Expenditure

With the development of single-use bioreactor devices that are well-automated, it has become possible to regulate the environment actively. Compared to traditional stainless-steel systems, it needs less upfront investment, has lower operating costs, and provides greater adaptability. These are offered from approx. 50L–25000L size (Jacquemart et al. 2016; Mf et al. 2021). A study on integrated continuous processing reported that about 30% cost savings can be achieved using disposable technologies w.r.t SS batch process, examples- wave, orbital shaken (OS), stirred tank and pneumatically mixed bioreactors (Schmidt 2022). There is low risk of contamination also with these systems. Single-use bioreactor (microcarrier-based) can be used in case of viral vectors and vaccines (Gallo-Ramírez et al. 2015; Ton et al. 2023).

Genetic engineering of therapeutic proteins

Proteins regulate and mediate the body's normal, everyday functions and metabolic processes. Disruption of the normal protein and the usual pathway of protein leads to significant disorders, and any error during protein production or processing (such as mutation or misfolding) leads to disruption of the normal proteins. However, Gupta et al. (2017) report that such diseases may be cured through the administration of mutant protein. Such proteins must be manufactured in factories to ensure an external supply. The production process and the synthesis of human origin protein in bacterial, fungal, insect, or mammalian cell hosts can be assisted by a variety of genetic engineering methods (Gupta and Shukla 2017a). Methods such as host cell engineering, expression construct optimization, and media generation have all been employed to increase the yield of treatments (Tihanyi and Nyitray 2020). In host cell engineering, gene expression is modified using prior knowledge of linked biological pathways. Several techniques have been developed to modify genes, such as gene overexpression, gene knock-in and knock-out, and the clustered regularly interspaced short palindromic repeats—CRISPER associated protein 9 (CRISPER-Cas9)-mediated knock-in approach (Dana 2018; Kim et al. 2020). Site-specific gene knock-out and on-site heterologous gene overexpression in C. reinhardtii using CRISPR-Cas9 was investigated in a study (Kim et al. 2020). Optimizing expression constructs often makes use of targeted genome integration approaches. Transposons systems have been proven to increase integration efficiency (Rajendra et al. 2017; Balasubramanian et al. 2018), and recombinant CHO cells with a high production ability of > 7 g/L were discovered after only 2–3 weeks of integration. Gene integration mediated by transposons is still in its infancy and needs extensive testing before it can be put to practical use (Tihanyi and Nyitray 2020). Ubiquitous chromatin-opening elements (UCOEs) have been identified as a potential tool for preventing gene silencing and maintaining high production levels (Neville et al. 2017). Site-independent, stable, and high-level expression of the transgenes is also possible with the help of Bacterial Artificial Chromosomes (BACs). Some genetic engineering technologies that can aid in the exogenous synthesis of therapeutic proteins include zinc finger nucleases (ZNFs), transcription activator-like effector nucleases (TALENs), and CRISPR-CAS9 (Kim and Kim 2014; Lee et al. 2015; Sakuma et al. 2015). Emerging methods for host cell genomics and targeted expression of genes include the detection of transcriptional hot zones (Gaidukov et al. 2018). The development of omics technologies facilitated the study of cellular and molecular features. Genomic, transcriptomic, proteomic, and other data sets shed new light on these and other domains. Multiplex secretome analysis was used to investigate the impact of host cell proteins on the generation of recombinant therapeutic proteins in a study by Kol et al. (2020) (Kol et al. 2020). Therapeutic protein and host expression research have benefited greatly from advances in genetic engineering and omics technology (Tihanyi and Nyitray 2020). All of these, however, need the use of complex and expensive high-tech tools, materials, and information technology.

Challenges associated with therapeutic protein production

Therapeutic proteins are a rapidly growing class of biologics that have revolutionized the treatment of various diseases, including cancer, autoimmune disorders, and infectious diseases. However, the production of these complex molecules presents numerous challenges that need to be addressed to ensure their safety, efficacy, and affordability. In this review, we will discuss some of the challenges associated with therapeutic protein production and possible solutions to overcome them. One of the greatest challenges in the use of therapeutic proteins was their fast degradation due to short half-lives. And because of their quick renal clearance and obvious enzymatic activity during systemic circulation, these therapeutic protein molecules have a short lifetime in blood. To compensate for this, a higher dose is usually required to maintain a desired concentration for a desired period in blood serum. So, a few technologies have been proposed and tested for the extension of half-life of therapeutic proteins such as amino acid manipulation, bio-conjugation/genetic fusion, post-translational modification by attaching the peptide to natural/synthetic polymers (PEGylation, polysialylation, HESylation) and carrier-mediated delivery (Tripathi and Shrivastava 2019). Another challenge of therapeutic protein production is the heterogeneity of the protein molecules. Unlike small molecules, which are chemically synthesized, therapeutic proteins are biologically produced in living cells, leading to variations in post-translational modifications, folding, and aggregation (Zhuang et al. 2023). This heterogeneity can affect the protein's stability, pharmacokinetics, and immunogenicity, leading to variability in efficacy and safety (Stebbings et al. 2009; Shih 2012; Jiskoot et al. 2012). To address this challenge, several approaches have been developed, including the use of advanced cell lines, optimized culture conditions, and novel purification techniques (Walsh 2018). Another significant challenge in therapeutic protein production is the high cost of production, which limits their accessibility to patients. The cost is driven by several factors, including the complexity of the manufacturing process, regulatory requirements, and intellectual property issues (Qarawi et al. 2019). To reduce the cost of production, various strategies have been explored, including process intensification, cell line engineering, and process optimization using advanced analytics and automation. Moreover, the production of therapeutic proteins also poses challenges related to scalability and manufacturing capacity (Chennamsetty et al. 2009; Hamid Akash et al. 2015). Many biologics are produced using mammalian cells, which require large-scale cell culture facilities and specialized equipment, making it challenging to produce enough proteins to meet the growing demand. Alternative expression systems, such as microbial cells and plant cells, have been explored to overcome this challenge (Buyel 2018).

Another challenge associated with protein therapeutics is immunogenicity. A modification in the protein's structure, which might happen as a result of post-translational changes such as product's administration, storage, or manufacturing process, sets off the immune response (Dingman and Balu-Iyer 2019). While a vaccine induced immune response is desired clinical response while immune response due to therapeutic proteins is undesirable. Today, a wide range of preclinical methods, including computational (HLA-binding algorithms) (Wang et al. 2008; De Groot et al. 2008; Chen et al. 2023), in vitro (Peptide/HLA-binding assay) (Justesen et al. 2009; Hamuro et al. 2017; Groell et al. 2018), ex vivo (LH/MS-based MHC-associated peptide proteomics, MHC-II tetramer-guided epitope mapping, protein-specific T-cell amplification, Human blood derived cell-based assay) (Bozzacco et al. 2011; Dudek et al. 2016) and animal models (HLA transgenic mice) (Jiskoot et al. 2016; Sauna et al. 2018), are employed to evaluate the immunogenicity risk of therapeutic proteins. The underlying assumption of most of these techniques is that the immunological reactions that are triggered by therapeutic proteins are dependent on T cells. A significant amount of research has demonstrated that the development of meaningful anti-drug antibodies (ADA) responses depends on T helper cells (Th cells: CD4 + T cells) responding to protein epitopes. Antigen-presenting cells (APCs) surface-expressed human leukocyte antigen (HLA) molecules (major histocompatibility complex class II, MHC-II) deliver these epitopes to Th cells (Matucci et al. 2019; Jawa et al. 2020; Ducret et al. 2022). Hence, it becomes prerequisite to identify potential HLA- restricted immunogenic epitopes because these are necessary conditions for eliciting an immune response to therapeutic proteins. Mitigating unwanted immune responses associated with therapeutic protein-based drug administration is crucial for disease management and cure. Some other approaches to reduce the induced immunogenicity include interfering lymphocytes activation and induction of regulatory B and T cells (Salazar-Fontana et al. 2017). Table 5 shows some of the immunogenicity-mitigating approaches used with therapeutic proteins.

Table 5.

Some immunogenicity-mitigating approaches with therapeutic proteins

S. no Approaches Details Examples References

Deimmunization of therapeutic protein

(PEGylation, fusion to polypeptides, reductive methylation, glycosylation and polysialylation)

Eliminating non-self-amino acid sequences to prevent triggering unintended immunological reactions

Antibody optimization by the reduction of non-germline amino acid sequences is one of the more recent mAb deimmunization techniques that have been more geared explicitly toward possibly immunogenic portions of the primary amino acid sequence

Immunotoxins (Diphtheria toxin, Pseudomonas exotoxin A and ricin toxin) Pastan et al. (2006), Cizeau et al. (2009), Sauna et al. (2018), Lin and Dinner (2019), Zinsli et al. (2021)
2 Immune Tolerance Induction (ITI) ITI regimens should be taken into consideration when a patient's immune system renders a life-saving medication ineffective and no other treatment alternatives, such as bypass therapy, are available

Immune responses to enzyme replacement therapy (ERT) in Pompe disease

Tolerogenic nanoparticles (tNPs) such as synthetic virus particles (SVPs) e.g., poly(lactidecoglycolide) (PGLA) encapsulated with immunomodulator rapamycin

Kishimoto et al. (2016), McCarthy et al. (2017), Mazor et al. (2018), King et al. (2018), Nakar and Shapiro (2019), Desai et al. (2020)
3 Targeting and Manipulating Dendritic cells To control ADA responses. The term "professional APCs" refers to dendritic cells (DCs). Initiating an immune response to a foreign antigen, serve as the first step Murine DCs treated with vitamin D3 can induce tolerogenic DCs in vivo Penna et al. (2007), Caminschi et al. (2012), Macri et al. (2016), Salazar-Fontana et al. (2017)
4 Cell depletion through Immunosuppression Negative selection leads to the reduction of auto-reactive cells, which is one of the innate mechanisms of central tolerance. Similar deletion processes are used in germinal centers to eliminate peripheral B cells that express auto-reactive antibodies Cyclophosphamide Witmer and Young (2013), Tie et al. (2022)

Protein engineering can improve protein expression and folding by modifying the protein sequence to enhance solubility, stability, and folding efficiency. This can be achieved by introducing amino acid substitutions, deletions, or additions (Zhu et al. 2017). For example, a study reported that the introduction of a disulfide bond in the Fc domain of a monoclonal antibody improved its folding and stability (Zeng et al. 2018). Cell line engineering can improve protein expression and folding by modifying the host cell to enhance protein secretion, reduce protein degradation, and improve PTMs. This can be achieved by modifying the cell's protein quality control machinery, such as chaperones and proteases, or by introducing genes for PTM enzymes (Luo and Lee 2013). For example, a study reported that the overexpression of an endoplasmic reticulum chaperone improved the folding and secretion of a therapeutic protein (McLaughlin and Vandenbroeck 2011; Luo and Lee 2013). Process optimization is another way by which we can improve protein yield and quality by optimizing the culture conditions, expression systems, and purification methods. This can be achieved by optimizing the pH, temperature, oxygen, and nutrient conditions during protein expression and purification (Shukla and Thömmes 2010; Choi and Geletu 2018) (Fig. 2).

Fig. 2.

Fig. 2

Summary showing challenges and possible solutions for production of therapeutic proteins. Challenges with therapeutic protein production shown in left side, and right side showed the strategies using to combat those challenges

Conclusion and future perspectives

To treat critical human infectious diseases, macromolecular protein therapeutics have been proven to be effective and helped in evolving the medicinal area. Due to the advent of recombinant DNA technology (RDT), a huge number of therapeutic proteins have made their way to pharmaceutical market plus their demand is rising day by day. The continuous improvement in the bioprocessing sector has led to the production of quality products. Using the prokaryotic and eukaryotic host expression systems, modern molecular biology is at front foot to produce therapeutic proteins which are of importance in various cell therapies. The future potential of such therapies is huge as ‘n’ number of proteins are produced by microorganisms. These proteins can also be used in combination with small molecular drugs to give synergistic benefits. For example, treatment of EGFR-positive colon cancer.

To summarize, it can be stated that the advancement in biotechnology has contributed a lot in reverberating the field. However, there are still some aspects that need to be worked upon. Such as in case of integrated therapeutic bioprocessing, sterility is a matter of concern, so a sterile barrier is required between upstream and downstream. Feedback control systems also need to be developed as some parameters of USP can affect DSP operation. Even after adding some formulations to protein systems, clinical safety and immunogenicity have been an issue which can lead to protein instability. Gene annotation is also a limitation even after the genome sequences of some microbes such as filamentous fungi are available. Therefore, it should enable genome-wide screening in fungi so as to identify the candidate genes and can surely provide sustainable solutions for multiple diseases worldwide. In conclusion, the production of therapeutic proteins presents numerous challenges that need to be addressed to ensure their safety, efficacy, and affordability. While many solutions have been proposed, there is still a need for continued innovation and collaboration among academia, industry, and regulatory agencies to overcome these challenges and make these life-saving therapies accessible to all.

Acknowledgements

The authors acknowledge Chandigarh University, Gharuan for providing infrastructure support to conduct the research work.

Declarations

Conflict of interest

The authors declare no conflict of interest.

References

  1. Adan Gökbulut A, Arslanoğlu A. Purification and biochemical characterization of an extracellular lipase from psychrotolerant Pseudomonas fluorescens KE38. Turk J Biol. 2013;37:538–546. doi: 10.3906/biy-1211-10. [DOI] [Google Scholar]
  2. Ahmad I, Rubbab T, Deeba F, Naqvi SMS. Optimization of E. coli culture conditions for efficient DNA uptake by electroporation. Turk J Biol. 2014;38:568–573. doi: 10.3906/biy-1311-60. [DOI] [Google Scholar]
  3. Ahmad M, Hirz M, Pichler H, Schwab H. Protein expression in Pichia pastoris: recent achievements and perspectives for heterologous protein production. Appl Microbiol Biotechnol. 2014;98:5301–5317. doi: 10.1007/s00253-014-5732-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ahmed H, Elzahab HA, Alswiai G. Purification of antioxidant protein isolated from Peganum harmala and its protective effect against CCl4 toxicity in rats. Turk J Biol. 2013 doi: 10.3906/biy-1110-29. [DOI] [Google Scholar]
  5. Akash MSH, Rehman K, Li N, et al. Sustained delivery of IL-1Ra from Pluronic F127-based thermosensitive gel prolongs its therapeutic potentials. Pharm Res. 2012;29:3475–3485. doi: 10.1007/s11095-012-0843-0. [DOI] [PubMed] [Google Scholar]
  6. Akash MSH, Shen Q, Rehman K, Chen S. Interleukin-1 receptor antagonist: a new therapy for type 2 diabetes mellitus. J Pharm Sci. 2012;101:1647–1658. doi: 10.1002/jps.23057. [DOI] [PubMed] [Google Scholar]
  7. Al Shaer D, Al Musaimi O, Albericio F, De La Torre BG. 2019 FDA TIDES (peptides and oligonucleotides) harvest. Pharmaceuticals. 2020;13:40. doi: 10.3390/ph13030040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Albericio F, Kruger HG. Therapeutic peptides. Future Med Chem. 2012;4:1527–1531. doi: 10.4155/fmc.12.94. [DOI] [PubMed] [Google Scholar]
  9. Aleem A, Akbar Samad AB, Vaqar S (2023) Emerging Variants of SARS-CoV-2 And Novel Therapeutics Against Coronavirus (COVID-19). In: StatPearls. StatPearls Publishing, Treasure Island [PubMed]
  10. Amawi H, Abu Deiab GI, Aljabali A, AA, et al. COVID-19 pandemic: an overview of epidemiology, pathogenesis, diagnostics and potential vaccines and therapeutics. Ther Deliv. 2020;11:245–268. doi: 10.4155/tde-2020-0035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Amiri S, Adibzadeh S, Ghanbari S, et al. CRISPR-interceded CHO cell line development approaches. Biotech Bioeng. 2023;120:865–902. doi: 10.1002/bit.28329. [DOI] [PubMed] [Google Scholar]
  12. Aravindan R, Viruthagiri T, Seenivasan A, Subhagar S. Microbial production and biomedical applications of lovastatin. Indian J Pharm Sci. 2008;70:701. doi: 10.4103/0250-474X.49087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Aris P, Wei Y, Mohamadzadeh M, Xia X. Griseofulvin: an updated overview of old and current knowledge. Molecules. 2022;27:7034. doi: 10.3390/molecules27207034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Baeshen NA, Baeshen MN, Sheikh A, et al. Cell factories for insulin production. Microb Cell Fact. 2014;13:141. doi: 10.1186/s12934-014-0141-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Baeshen MN, Al-Hejin AM, Bora RS, et al. Production of biopharmaceuticals in E. coli: current scenario and future perspectives. J Microbiol Biotechnol. 2015;25:953–962. doi: 10.4014/jmb.1412.12079. [DOI] [PubMed] [Google Scholar]
  16. Baghban R, Farajnia S, Rajabibazl M, et al. Yeast expression systems: overview and recent advances. Mol Biotechnol. 2019;61:365–384. doi: 10.1007/s12033-019-00164-8. [DOI] [PubMed] [Google Scholar]
  17. Baghban R, Farajnia S, Ghasemi Y, et al. Mutational analysis of ocriplasmin to reduce proteolytic and autolytic activity in Pichia pastoris. Biol Proced Online. 2020;22:25. doi: 10.1186/s12575-020-00138-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bailon P, Won C-Y. PEG-modified biopharmaceuticals. Expert Opin Drug Deliv. 2009;6:1–16. doi: 10.1517/17425240802650568. [DOI] [PubMed] [Google Scholar]
  19. Balamurugan V, Reddy GR, Suryanarayana VV. Pichia pastoris: a notable heterologous expression system for the production of foreign proteins—vaccines. Ind J Biotechnol. 2007;6:175–186. [Google Scholar]
  20. Balasubramanian S, Peery RB, Minshull J, et al. Generation of high expressing chinese hamster ovary cell pools using the leap-in transposon system. Biotechnol J. 2018;13:1700748. doi: 10.1002/biot.201700748. [DOI] [PubMed] [Google Scholar]
  21. Bandaranayake AD, Almo SC. Recent advances in mammalian protein production. FEBS Lett. 2014;588:253–260. doi: 10.1016/j.febslet.2013.11.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Banerjee A, Ward V. Production of recombinant and therapeutic proteins in microalgae. Curr Opin Biotechnol. 2022;78:102784. doi: 10.1016/j.copbio.2022.102784. [DOI] [PubMed] [Google Scholar]
  23. Barahimipour R, Neupert J, Bock R. Efficient expression of nuclear transgenes in the green alga Chlamydomonas: synthesis of an HIV antigen and development of a new selectable marker. Plant Mol Biol. 2016;90:403–418. doi: 10.1007/s11103-015-0425-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Barbosa MJ, Janssen M, Südfeld C, et al. Hypes, hopes, and the way forward for microalgal biotechnology. Trends Biotechnol. 2023;41:452–471. doi: 10.1016/j.tibtech.2022.12.017. [DOI] [PubMed] [Google Scholar]
  25. Barman P, Joshi S, Sharma S, et al. Strategic approaches to improvise peptide drugs as next generation therapeutics. Int J Pept Res Ther. 2023;29:61. doi: 10.1007/s10989-023-10524-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bayry J, Aimanianda V, Guijarro JI, et al. Hydrophobins—unique fungal proteins. PLoS Pathog. 2012;8:e1002700. doi: 10.1371/journal.ppat.1002700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Belén LH, Beltrán Lissabet JF, De Oliveira R-Y, et al. Immunogenicity assessment of fungal l-asparaginases: an in silico approach. SN Appl Sci. 2020;2:222. doi: 10.1007/s42452-020-2021-z. [DOI] [Google Scholar]
  28. Berndt AJ, Smalley TN, Ren B, et al. Recombinant production of a functional SARS-CoV-2 spike receptor binding domain in the green algae Chlamydomonas reinhardtii. PLoS ONE. 2021;16:e0257089. doi: 10.1371/journal.pone.0257089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bertolini LR, Meade H, Lazzarotto CR, et al. The transgenic animal platform for biopharmaceutical production. Transgenic Res. 2016;25:329–343. doi: 10.1007/s11248-016-9933-9. [DOI] [PubMed] [Google Scholar]
  30. Bhatia S, Singh A, Batra N, Singh J. Microbial production and biotechnological applications of α-galactosidase. Int J Biol Macromol. 2020;150:1294–1313. doi: 10.1016/j.ijbiomac.2019.10.140. [DOI] [PubMed] [Google Scholar]
  31. Bhuvanesh S, Arunkumar C, Kaliraj P, Ramalingam S. Production and single-step purification of Brugia malayi abundant larval transcript (ALT-2) using hydrophobic interaction chromatography. J Ind Microbiol Biotechnol. 2010;37:1053–1059. doi: 10.1007/s10295-010-0753-2. [DOI] [PubMed] [Google Scholar]
  32. Bill RM. Recombinant protein subunit vaccine synthesis in microbes: a role for yeast? J Pharm Pharmacol. 2015;67:319–328. doi: 10.1111/jphp.12353. [DOI] [PubMed] [Google Scholar]
  33. Borovsky D, Sterner A, Powell CA. Cloning and expressing trypsin modulating oostatic factor in Chlorella desiccata to control mosquito larvae: expression of TMOF in Chlorella desiccata to control mosquito larvae. Arch Insect Biochem Physiol. 2016;91:17–36. doi: 10.1002/arch.21306. [DOI] [PubMed] [Google Scholar]
  34. Bozzacco L, Yu H, Zebroski HA, et al. Mass spectrometry analysis and quantitation of peptides presented on the MHC II molecules of mouse spleen dendritic cells. J Proteome Res. 2011;10:5016–5030. doi: 10.1021/pr200503g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Buchwald H, Dorman RB, Rasmus NF, et al. Effects on GLP-1, PYY, and leptin by direct stimulation of terminal ileum and cecum in humans: implications for ileal transposition. Surg Obes Related Dis. 2014;10:780–786. doi: 10.1016/j.soard.2014.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Burnett MJB, Burnett AC. Therapeutic recombinant protein production in plants: challenges and opportunities. Plants People Planet. 2020;2:121–132. doi: 10.1002/ppp3.10073. [DOI] [Google Scholar]
  37. Buyel JF. Plants as sources of natural and recombinant anti-cancer agents. Biotechnol Adv. 2018;36:506–520. doi: 10.1016/j.biotechadv.2018.02.002. [DOI] [PubMed] [Google Scholar]
  38. Buyel JF, Twyman RM, Fischer R. Very-large-scale production of antibodies in plants: the biologization of manufacturing. Biotechnol Adv. 2017;35:458–465. doi: 10.1016/j.biotechadv.2017.03.011. [DOI] [PubMed] [Google Scholar]
  39. Caetano KAA, Del-Rios NHA, Pinheiro RS, et al. Low immunogenicity of recombinant hepatitis b vaccine derived from Hansenula polymorpha in adults aged over 40 years. Am J Trop Med Hyg. 2017;96:118–121. doi: 10.4269/ajtmh.16-0475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Cai D, Wang H, He P, et al. A novel strategy to improve protein secretion via overexpression of the SppA signal peptide peptidase in Bacillus licheniformis. Microb Cell Fact. 2017;16:70. doi: 10.1186/s12934-017-0688-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Caminschi I, Maraskovsky E, Heath WR. Targeting dendritic cells in vivo for cancer therapy. Front Immunol. 2012;3:13. doi: 10.3389/fimmu.2012.00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Cammalleri L, Malaguarnera M. Rasburicase represents a new tool for hyperuricemia in tumor lysis syndrome and in gout. Int J Med Sci. 2007 doi: 10.7150/ijms.4.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Campos-Neto A, Rodrigues-Júnior V, Pedral-Sampaio DB, et al. Evaluation of DPPD, a single recombinant Mycobacterium tuberculosis protein as an alternative antigen for the Mantoux test. Tuberculosis. 2001;81:353–358. doi: 10.1054/tube.2001.0311. [DOI] [PubMed] [Google Scholar]
  44. Cao J, De La Fuente-Nunez C, Ou RW, et al. Yeast-based synthetic biology platform for antimicrobial peptide production. ACS Synth Biol. 2018;7:896–902. doi: 10.1021/acssynbio.7b00396. [DOI] [PubMed] [Google Scholar]
  45. Celińska E, Białas W, Borkowska M, Grajek W. Cloning, expression, and purification of insect (Sitophilus oryzae) alpha-amylase, able to digest granular starch, in Yarrowia lipolytica host. Appl Microbiol Biotechnol. 2015;99:2727–2739. doi: 10.1007/s00253-014-6314-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Celińska E, Borkowska M, Białas W. Evaluation of heterologous α -amylase production in two expression platforms dedicated for Yarrowia lipolytica : commercial Po1g-pYLSC ( php4d ) and custom-made A18-pYLTEF ( pTEF ): comparison of two Yarrowia -based systems in expression of α -amylase. Yeast. 2016;33:165–181. doi: 10.1002/yea.3149. [DOI] [PubMed] [Google Scholar]
  47. Celińska E, Borkowska M, Białas W. Enhanced production of insect raw-starch-digesting alpha-amylase accompanied by high erythritol synthesis in recombinant Yarrowia lipolytica fed-batch cultures at high-cell-densities. Process Biochem. 2017;52:78–85. doi: 10.1016/j.procbio.2016.10.022. [DOI] [Google Scholar]
  48. Chaudet MM, Allen J-L, Rose DR. Expression and purification of two Family GH31 α-glucosidases from Bacteroides thetaiotaomicron. Protein Expr Purif. 2012;86:135–141. doi: 10.1016/j.pep.2012.09.009. [DOI] [PubMed] [Google Scholar]
  49. Chen L, Yang X, Luo D, Yu W. Efficient production of a bioactive bevacizumab monoclonal antibody using the 2a self-cleavage peptide in transgenic rice callus. Front Plant Sci. 2016 doi: 10.3389/fpls.2016.01156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Chen B-M, Cheng T-L, Roffler SR. Polyethylene glycol immunogenicity: theoretical, clinical, and practical aspects of anti-polyethylene glycol antibodies. ACS Nano. 2021;15:14022–14048. doi: 10.1021/acsnano.1c05922. [DOI] [PubMed] [Google Scholar]
  51. Chen Z, Wang X, Chen X, et al. Accelerating therapeutic protein design with computational approaches toward the clinical stage. Comput Struct Biotechnol J. 2023;21:2909–2926. doi: 10.1016/j.csbj.2023.04.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Chennamsetty N, Voynov V, Kayser V, et al. Design of therapeutic proteins with enhanced stability. Proc Natl Acad Sci USA. 2009;106:11937–11942. doi: 10.1073/pnas.0904191106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Choi T-J, Geletu TT. High level expression and purification of recombinant flounder growth hormone in E. coli. J Genet Eng Biotechnol. 2018;16:347–355. doi: 10.1016/j.jgeb.2018.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Chung M-E, Yeh I-H, Sung L-Y, et al. Enhanced integration of large DNA into E. coli chromosome by CRISPR/Cas9: CRISPR/Cas9 for bacterial genome engineering. Biotechnol Bioeng. 2017;114:172–183. doi: 10.1002/bit.26056. [DOI] [PubMed] [Google Scholar]
  55. Cizeau J, Grenkow DM, Brown JG, et al. Engineering and biological characterization of VB6-845, an anti-EpCAM immunotoxin containing a T-cell epitope-depleted variant of the plant toxin bouganin. J Immunother. 2009;32:574–584. doi: 10.1097/CJI.0b013e3181a6981c. [DOI] [PubMed] [Google Scholar]
  56. Clausen H, Wandall HH, DeLisa MP, Stanley P, Schnaar RL. Essentials of glycobiology. 4. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press; 2022. Glycosylation engineering. [Google Scholar]
  57. Coler RN, Skeiky YAW, Ovendale PJ, et al. Cloning of a Mycobacterium tuberculosis gene encoding a purifed protein derivative protein that elicits strong tuberculosis-specific delayed-type hypersensitivity. J INFECT DIS. 2000;182:224–233. doi: 10.1086/315677. [DOI] [PubMed] [Google Scholar]
  58. Commault AS, Kaur Walia N, Fabris M, et al. Effect of biphasic temperature regime on therapeutic recombinant protein production in the green alga Chlamydomonas reinhardtii. Algal Res. 2020;50:101997. doi: 10.1016/j.algal.2020.101997. [DOI] [Google Scholar]
  59. Corwin HL, Gettinger A, Pearl RG, et al. Efficacy of recombinant human erythropoietin in critically ill patients: a randomized controlled trial. JAMA. 2002;288:2827. doi: 10.1001/jama.288.22.2827. [DOI] [PubMed] [Google Scholar]
  60. Craig TJ, Li HH, Riedl M, et al. Characterization of anaphylaxis after ecallantide treatment of hereditary angioedema attacks. J Allergy Clin Immunol In Pract. 2015;3:206–212.e4. doi: 10.1016/j.jaip.2014.09.001. [DOI] [PubMed] [Google Scholar]
  61. Craik DJ, Fairlie DP, Liras S, Price D. The future of peptide-based drugs. Chem Biol Drug Des. 2013;81:136–147. doi: 10.1111/cbdd.12055. [DOI] [PubMed] [Google Scholar]
  62. Crosnier J, Jungers P, Couroucé A, et al. Randomised placebo-controlled trial of hepatitis B surface antigen vaccine in French haemodialysis units: II, haemodialysis patients. The Lancet. 1981;317:797–800. doi: 10.1016/S0140-6736(81)92679-9. [DOI] [PubMed] [Google Scholar]
  63. Dana R. Advances and innovations in recombinant protein expression technology. J Adv Pharm Educ Res. 2018;8:89. [Google Scholar]
  64. Darvishi F, Moradi M, Madzak C, Jolivalt C. Production of laccase by recombinant Yarrowia lipolytica from molasses: bioprocess development using statistical modeling and increase productivity in shake-flask and bioreactor cultures. Appl Biochem Biotechnol. 2017;181:1228–1239. doi: 10.1007/s12010-016-2280-8. [DOI] [PubMed] [Google Scholar]
  65. Das S, Al-Toubah T, El-Haddad G, Strosberg J. 177Lu-DOTATATE for the treatment of gastroenteropancreatic neuroendocrine tumors. Expert Rev Gastroenterol Hepatol. 2019;13:1023–1031. doi: 10.1080/17474124.2019.1685381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. De Jesus M, Wurm FM. Manufacturing recombinant proteins in kg-ton quantities using animal cells in bioreactors. Eur J Pharm Biopharm. 2011;78:184–188. doi: 10.1016/j.ejpb.2011.01.005. [DOI] [PubMed] [Google Scholar]
  67. De Groot AS, McMurry J, Moise L. Prediction of immunogenicity: in silico paradigms, ex vivo and in vivo correlates. Curr Opin Pharmacol. 2008;8:620–626. doi: 10.1016/j.coph.2008.08.002. [DOI] [PubMed] [Google Scholar]
  68. Dehghani J, Adibkia K, Movafeghi A, et al (2020) Designing a new generation of expression toolkits for engineering of green microalgae; robust production of human interleukin-2. Bioimpacts 10:259–268. 10.34172/bi.2020.33 [DOI] [PMC free article] [PubMed]
  69. Demain AL, Vaishnav P. Production of recombinant proteins by microbes and higher organisms. Biotechnol Adv. 2009;27:297–306. doi: 10.1016/j.biotechadv.2009.01.008. [DOI] [PubMed] [Google Scholar]
  70. Demurtas OC, Massa S, Ferrante P, et al. A chlamydomonas-derived human papillomavirus 16 E7 vaccine induces specific tumor protection. PLoS ONE. 2013;8:e61473. doi: 10.1371/journal.pone.0061473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Deniz I, García-Vaquero M, Imamoglu E. Microalgae-based biofuels and bioproducts. New York: Elsevier; 2017. Trends in red biotechnology; pp. 429–460. [Google Scholar]
  72. Desai AK, Baloh CH, Sleasman JW, et al. Benefits of prophylactic short-course immune tolerance induction in patients with infantile pompe disease: demonstration of long-term safety and efficacy in an expanded cohort. Front Immunol. 2020;11:1727. doi: 10.3389/fimmu.2020.01727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Dingman R, Balu-Iyer SV. Immunogenicity of protein pharmaceuticals. J Pharm Sci. 2019;108:1637–1654. doi: 10.1016/j.xphs.2018.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Dirisala VR, Nair RR, Srirama K, et al. Recombinant pharmaceutical protein production in plants: unraveling the therapeutic potential of molecular pharming. Acta Physiol Plant. 2017;39:18. doi: 10.1007/s11738-016-2315-3. [DOI] [Google Scholar]
  75. Donini M, Marusic C. Current state-of-the-art in plant-based antibody production systems. Biotechnol Lett. 2019;41:335–346. doi: 10.1007/s10529-019-02651-z. [DOI] [PubMed] [Google Scholar]
  76. Du T, Buenbrazo N, Kell L, et al. Cell Chem Biol. 2019;26:203–212.e5. doi: 10.1016/j.chembiol.2018.10.017. [DOI] [PubMed] [Google Scholar]
  77. Du L, Yang Y, Zhang X. Neutralizing antibodies for the prevention and treatment of COVID-19. Cell Mol Immunol. 2021;18:2293–2306. doi: 10.1038/s41423-021-00752-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Duchin JS, Jereb JA, Nolan CM, et al. Comparison of sensitivities to two commercially available tuberculin skin test reagents in persons with recent tuberculosis. CLIN INFECT DIS. 1997;25:661–663. doi: 10.1086/513771. [DOI] [PubMed] [Google Scholar]
  79. Ducret A, Ackaert C, Bessa J, et al Assay format diversity in pre-clinical immunogenicity risk assessment: toward a possible harmonization of antigenicity assays. MAbs 14:1993522. 10.1080/19420862.2021.1993522 [DOI] [PMC free article] [PubMed]
  80. Dudek NL, Croft NP, Schittenhelm RB, et al. A systems approach to understand antigen presentation and the immune response. Methods Mol Biol. 2016;1394:189–209. doi: 10.1007/978-1-4939-3341-9_14. [DOI] [PubMed] [Google Scholar]
  81. Dumont J, Euwart D, Mei B, et al. Human cell lines for biopharmaceutical manufacturing: history, status, and future perspectives. Crit Rev Biotechnol. 2016;36:1110–1122. doi: 10.3109/07388551.2015.1084266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Dyson MR. Fundamentals of expression in mammalian cells. In: Vega MC, editor. Advanced technologies for protein complex production and characterization. Cham: Springer International Publishing; 2016. pp. 217–224. [Google Scholar]
  83. Eichler-Stahlberg A, Weisheit W, Ruecker O, Heitzer M. Strategies to facilitate transgene expression in Chlamydomonas reinhardtii. Planta. 2009;229:873–883. doi: 10.1007/s00425-008-0879-x. [DOI] [PubMed] [Google Scholar]
  84. El-Gendy MMAA, Awad MF, El-Shenawy FS, El-Bondkly AMA. Production, purification, characterization, antioxidant and antiproliferative activities of extracellular L-asparaginase produced by Fusarium equiseti AHMF4. Saudi J Biol Sci. 2021;28:2540–2548. doi: 10.1016/j.sjbs.2021.01.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Eriksson BI, Wille-Jørgensen P, Kälebo P, et al. A comparison of recombinant hirudin with a low-molecular-weight heparin to prevent thromboembolic complications after total hip replacement. N Engl J Med. 1997;337:1329–1335. doi: 10.1056/NEJM199711063371901. [DOI] [PubMed] [Google Scholar]
  86. Evangelista L, Ravelli I, Bignotto A, et al. Ga-68 DOTA-peptides and F-18 FDG PET/CT in patients with neuroendocrine tumor: a review. Clin Imaging. 2020;67:113–116. doi: 10.1016/j.clinimag.2020.05.035. [DOI] [PubMed] [Google Scholar]
  87. Fahad S, Khan FA, Pandupuspitasari NS, et al. Recent developments in therapeutic protein expression technologies in plants. Biotechnol Lett. 2015;37:265–279. doi: 10.1007/s10529-014-1699-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Fan L, Kadura I, Krebs LE, et al. Development of a highly-efficient CHO cell line generation system with engineered SV40E promoter. J Biotechnol. 2013;168:652–658. doi: 10.1016/j.jbiotec.2013.08.021. [DOI] [PubMed] [Google Scholar]
  89. Fernandes D, Pacheco LK, Sordi R, et al. Angiotensin II receptor type 1 blockade improves hyporesponsiveness to vasopressors in septic shock. Eur J Pharmacol. 2021;897:173953. doi: 10.1016/j.ejphar.2021.173953. [DOI] [PubMed] [Google Scholar]
  90. Ferrer-Miralles N, Domingo-Espín J, Corchero JL, et al. Microbial factories for recombinant pharmaceuticals. Microb Cell Fact. 2009;8:17. doi: 10.1186/1475-2859-8-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Fink M, Cserjan-Puschmann M, Reinisch D, Striedner G. High-throughput microbioreactor provides a capable tool for early stage bioprocess development. Sci Rep. 2021;11:2056. doi: 10.1038/s41598-021-81633-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Fischer R, Holland T, Sack M, et al. Glyco-engineering of plant-based expression systems. In: Rapp E, Reichl U, et al., editors. Advances in glycobiotechnology. Cham: Springer International Publishing; 2018. pp. 137–166. [DOI] [PubMed] [Google Scholar]
  93. Fisher AC, Kamga M-H, Agarabi C, et al. The current scientific and regulatory landscape in advancing integrated continuous biopharmaceutical manufacturing. Trends Biotechnol. 2019;37:253–267. doi: 10.1016/j.tibtech.2018.08.008. [DOI] [PubMed] [Google Scholar]
  94. Fletcher E, Krivoruchko A, Nielsen J. Industrial systems biology and its impact on synthetic biology of yeast cell factories: systems Biology and It’s Impact on Synthetic Biology. Biotechnol Bioeng. 2016;113:1164–1170. doi: 10.1002/bit.25870. [DOI] [PubMed] [Google Scholar]
  95. Fosgerau K, Hoffmann T. Peptide therapeutics: current status and future directions. Drug Discovery Today. 2015;20:122–128. doi: 10.1016/j.drudis.2014.10.003. [DOI] [PubMed] [Google Scholar]
  96. Friedl C, Zitt E. Role of etelcalcetide in the management of secondary hyperparathyroidism in hemodialysis patients: a review on current data and place in therapy. Drug Des Devel Ther. 2018;12:1589–1598. doi: 10.2147/DDDT.S134103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Fu W, Chaiboonchoe A, Khraiwesh B, et al. Algal cell factories: approaches, applications, and potentials. Mar Drugs. 2016;14:225. doi: 10.3390/md14120225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Fu W, Nelson DR, Mystikou A, et al. Advances in microalgal research and engineering development. Curr Opin Biotechnol. 2019;59:157–164. doi: 10.1016/j.copbio.2019.05.013. [DOI] [PubMed] [Google Scholar]
  99. Fukuda N, Senga Y, Honda S (2019) Anxa2—and Ctsd‐knockout CHO cell lines to diminish the risk of contamination with host cell proteins. Biotechnol Progr. 10.1002/btpr.2820 [DOI] [PubMed]
  100. Furman BL (2017) Aspart Insulin. In: Reference module in biomedical sciences. Elsevier, New York
  101. Gaidukov L, Wroblewska L, Teague B, et al. A multi-landing pad DNA integration platform for mammalian cell engineering. Nucleic Acids Res. 2018;46:4072–4086. doi: 10.1093/nar/gky216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Gallo-Ramírez LE, Nikolay A, Genzel Y, Reichl U. Bioreactor concepts for cell culture-based viral vaccine production. Expert Rev Vaccines. 2015;14:1181–1195. doi: 10.1586/14760584.2015.1067144. [DOI] [PubMed] [Google Scholar]
  103. Garcia JM, Biller BMK, Korbonits M, et al. Sensitivity and specificity of the macimorelin test for diagnosis of AGHD. Endocr Connect. 2021;10:76–83. doi: 10.1530/EC-20-0491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Gasmi N, Lassoued R, Ayed A, et al. Production and characterization of human granulocyte–macrophage colony-stimulating factor (hGM-CSF) expressed in the oleaginous yeast Yarrowia lipolytica. Appl Microbiol Biotechnol. 2012;96:89–101. doi: 10.1007/s00253-012-4141-x. [DOI] [PubMed] [Google Scholar]
  105. Gellissen G, Melber K, Janowicz ZA, et al. Heterologous protein production in yeast. Antonie Van Leeuwenhoek. 1992;62:79–93. doi: 10.1007/BF00584464. [DOI] [PubMed] [Google Scholar]
  106. Ghaderi D, Taylor RE, Padler-Karavani V, et al. Implications of the presence of N-glycolylneuraminic acid in recombinant therapeutic glycoproteins. Nat Biotechnol. 2010;28:863–867. doi: 10.1038/nbt.1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Ghaderi D, Zhang M, Hurtado-Ziola N, Varki A. Production platforms for biotherapeutic glycoproteins. Occurrence, impact, and challenges of non-human sialylation. Biotechnol Genet Eng Rev. 2012;28:147–176. doi: 10.5661/bger-28-147. [DOI] [PubMed] [Google Scholar]
  108. Giordano C, Marchiò M, Timofeeva E, Biagini G. Neuroactive peptides as putative mediators of antiepileptic ketogenic diets. Front Neurol. 2014;5:63. doi: 10.3389/fneur.2014.00063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Gomaa EZ. Production, characterization, and antitumor efficiency of l-glutaminase from halophilic bacteria. Bull Natl Res Cent. 2022;46:10. doi: 10.1186/s42269-021-00693-w. [DOI] [Google Scholar]
  110. Groell F, Jordan O, Borchard G. In vitro models for immunogenicity prediction of therapeutic proteins. Eur J Pharm Biopharm. 2018;130:128–142. doi: 10.1016/j.ejpb.2018.06.008. [DOI] [PubMed] [Google Scholar]
  111. Gronemeyer P, Ditz R, Strube J. Trends in upstream and downstream process development for antibody manufacturing. Bioengineering. 2014;1:188–212. doi: 10.3390/bioengineering1040188. [DOI] [PubMed] [Google Scholar]
  112. Gupta SK, Dangi AK, Smita M, et al (2019) Effectual bioprocess development for protein production. In: Applied microbiology and bioengineering. Elsevier, New York, pp 203–227
  113. Gupta V, Sengupta M, Prakash J, Tripathy BC, Gupta V, Sengupta M, Prakash J, Tripathy BC. Production of recombinant pharmaceutical proteins. Basic Appl Asp Biotechnol. 2017 doi: 10.1007/978-981-10-0875-7_4. [DOI] [Google Scholar]
  114. Gupta SK, Shukla P. Advanced technologies for improved expression of recombinant proteins in bacteria: perspectives and applications. Crit Rev Biotechnol. 2016;36:1089–1098. doi: 10.3109/07388551.2015.1084264. [DOI] [PubMed] [Google Scholar]
  115. Gupta SK, Shukla P. Gene editing for cell engineering: trends and applications. Crit Rev Biotechnol. 2017;37:672–684. doi: 10.1080/07388551.2016.1214557. [DOI] [PubMed] [Google Scholar]
  116. Gupta SK, Shukla P. Sophisticated cloning, fermentation, and purification technologies for an enhanced therapeutic protein production: a review. Front Pharmacol. 2017;8:419. doi: 10.3389/fphar.2017.00419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Hamid Akash MS, Rehman K, Chen S. Natural and synthetic polymers as drug carriers for delivery of therapeutic proteins. Polym Rev. 2015;55:371–406. doi: 10.1080/15583724.2014.995806. [DOI] [Google Scholar]
  118. Hamuro L, Kijanka G, Kinderman F, et al. Perspectives on subcutaneous route of administration as an immunogenicity risk factor for therapeutic proteins. J Pharm Sci. 2017;106:2946–2954. doi: 10.1016/j.xphs.2017.05.030. [DOI] [PubMed] [Google Scholar]
  119. Hanke AT, Ottens M. Purifying biopharmaceuticals: knowledge-based chromatographic process development. Trends Biotechnol. 2014;32:210–220. doi: 10.1016/j.tibtech.2014.02.001. [DOI] [PubMed] [Google Scholar]
  120. Haws R, Brady S, Davis E, et al. Effect of setmelanotide, a melanocortin-4 receptor agonist, on obesity in Bardet-Biedl syndrome. Diabetes Obes Metab. 2020;22:2133–2140. doi: 10.1111/dom.14133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Hayashi K, Lee J-B, Atsumi K, et al (2019) In vitro and in vivo anti-herpes simplex virus activity of monogalactosyl diacylglyceride from Coccomyxa sp. KJ (IPOD FERM BP-22254), a green microalga. PLoS ONE 14:e0219305. 10.1371/journal.pone.0219305 [DOI] [PMC free article] [PubMed]
  122. Hefferon K (2017) Reconceptualizing cancer immunotherapy based on plant production systems. Future Sci OA 3:FSO217. 10.4155/fsoa-2017-0018 [DOI] [PMC free article] [PubMed]
  123. Heffner KM, Wang Q, Hizal DB, et al. Glycoengineering of mammalian expression systems on a cellular level. In: Rapp E, Reichl U, et al., editors. Advances in glycobiotechnology. Cham: Springer International Publishing; 2018. pp. 37–69. [DOI] [PubMed] [Google Scholar]
  124. Hempel F, Lau J, Klingl A, Maier UG. Algae as protein factories: expression of a human antibody and the respective antigen in the diatom Phaeodactylum tricornutum. PLoS ONE. 2011;6:e28424. doi: 10.1371/journal.pone.0028424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Hempel F, Maurer M, Brockmann B, et al. From hybridomas to a robust microalgal-based production platform: molecular design of a diatom secreting monoclonal antibodies directed against the Marburg virus nucleoprotein. Microb Cell Fact. 2017;16:131. doi: 10.1186/s12934-017-0745-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Henninot A, Collins JC, Nuss JM. The current state of peptide drug discovery: back to the future? J Med Chem. 2018;61:1382–1414. doi: 10.1021/acs.jmedchem.7b00318. [DOI] [PubMed] [Google Scholar]
  127. Hernández-Ramírez J, Wong-Arce A, González-Ortega O, Rosales-Mendoza S. Expression in algae of a chimeric protein carrying several epitopes from tumor associated antigens. Int J Biol Macromol. 2020;147:46–52. doi: 10.1016/j.ijbiomac.2019.12.250. [DOI] [PubMed] [Google Scholar]
  128. Holtz BR, Berquist BR, Bennett LD, et al. Commercial-scale biotherapeutics manufacturing facility for plant-made pharmaceuticals. Plant Biotechnol J. 2015;13:1180–1190. doi: 10.1111/pbi.12469. [DOI] [PubMed] [Google Scholar]
  129. Hu J, Han J, Li H, et al. Human embryonic kidney 293 cells: a vehicle for biopharmaceutical manufacturing, structural biology, and electrophysiology. Cells Tissues Organs. 2018;205:1–8. doi: 10.1159/000485501. [DOI] [PubMed] [Google Scholar]
  130. Huang C, Ding T, Wang J, et al. CRISPR-Cas9-assisted native end-joining editing offers a simple strategy for efficient genetic engineering in Escherichia coli. Appl Microbiol Biotechnol. 2019;103:8497–8509. doi: 10.1007/s00253-019-10104-w. [DOI] [PubMed] [Google Scholar]
  131. Huertas MJ, Michán C. Paving the way for the production of secretory proteins by yeast cell factories. Microb Biotechnol. 2019;12:1095–1096. doi: 10.1111/1751-7915.13342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Hunter M, Yuan P, Vavilala D, Fox M. Optimization of protein expression in mammalian cells. Curr Protoc Protein Sci. 2019;95:e77. doi: 10.1002/cpps.77. [DOI] [PubMed] [Google Scholar]
  133. Ingle KP, Singh A, Sahni T, et al (2023) Genome editing in the synthetic biology for sustainable production of biomolecules. In: Singh V, Show PL (eds) Biomanufacturing for sustainable production of biomolecules. Springer Nature Singapore, Singapore, pp 315–329
  134. Jacquemart R, Vandersluis M, Zhao M, et al. A single-use strategy to enable manufacturing of affordable biologics. Comput Struct Biotechnol J. 2016;14:309–318. doi: 10.1016/j.csbj.2016.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Jarquín-Cordero M, Chávez MN, Centeno-Cerdas C, et al. Towards a biotechnological platform for the production of human pro-angiogenic growth factors in the green alga Chlamydomonas reinhardtii. Appl Microbiol Biotechnol. 2020;104:725–739. doi: 10.1007/s00253-019-10267-6. [DOI] [PubMed] [Google Scholar]
  136. Jawa V, Terry F, Gokemeijer J, et al. T-cell dependent immunogenicity of protein therapeutics pre-clinical assessment and mitigation-updated consensus and review 2020. Front Immunol. 2020;11:1301. doi: 10.3389/fimmu.2020.01301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Jazayeri SH, Amiri-Yekta A, Bahrami S, et al. Vector and cell line engineering technologies toward recombinant protein expression in mammalian cell lines. Appl Biochem Biotechnol. 2018;185:986–1003. doi: 10.1007/s12010-017-2689-8. [DOI] [PubMed] [Google Scholar]
  138. Jesuraj SAV, Sarker MdMR, Ming LC, et al. Enhancement of the production of L-glutaminase, an anticancer enzyme, from Aeromonas veronii by adaptive and induced mutation techniques. PLoS ONE. 2017;12:e0181745. doi: 10.1371/journal.pone.0181745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Ji X, Peng X, Long X, et al. Laccase-mediated functionalization of natamycin by gallic acids for the therapeutic effect on Aspergillus fumigatus keratitis. Eur J Pharmacol. 2022;926:175041. doi: 10.1016/j.ejphar.2022.175041. [DOI] [PubMed] [Google Scholar]
  140. Jiskoot W, Randolph TW, Volkin DB, et al. Protein instability and immunogenicity: roadblocks to clinical application of injectable protein delivery systems for sustained release. J Pharm Sci. 2012;101:946–954. doi: 10.1002/jps.23018. [DOI] [PubMed] [Google Scholar]
  141. Jiskoot W, Kijanka G, Randolph TW, et al. Mouse models for assessing protein immunogenicity: lessons and challenges. J Pharm Sci. 2016;105:1567–1575. doi: 10.1016/j.xphs.2016.02.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Jozala AF, Geraldes DC, Tundisi LL, et al. Biopharmaceuticals from microorganisms: from production to purification. Braz J Microbiol. 2016;47:51–63. doi: 10.1016/j.bjm.2016.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Justesen S, Harndahl M, Lamberth K, et al. Functional recombinant MHC class II molecules and high-throughput peptide-binding assays. Immunome Res. 2009;5:2. doi: 10.1186/1745-7580-5-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Juturu V, Wu JC. Heterologous protein expression in Pichia pastoris : latest research progress and applications. ChemBioChem. 2018;19:7–21. doi: 10.1002/cbic.201700460. [DOI] [PubMed] [Google Scholar]
  145. Kaewput C, Vinjamuri S. Role of combined 68Ga DOTA-peptides and 18F FDG PET/CT in the evaluation of gastroenteropancreatic neuroendocrine neoplasms. Diagnostics (Basel) 2022;12:280. doi: 10.3390/diagnostics12020280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Kamermans A, Verhoeven T, van het Hof B, et al. Setmelanotide, a novel, selective melanocortin receptor-4 agonist exerts anti-inflammatory actions in astrocytes and promotes an anti-inflammatory macrophage phenotype. Front Immunol. 2019;10:2312. doi: 10.3389/fimmu.2019.02312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Kamionka M (2011) Engineering of therapeutic proteins production in Escherichia coli. curr pharm biotechnol 12:268–274. 10.2174/138920111794295693 [DOI] [PMC free article] [PubMed]
  148. Kasli IM, Thomas ORT, Overton TW. Use of a design of experiments approach to optimise production of a recombinant antibody fragment in the periplasm of Escherichia coli: selection of signal peptide and optimal growth conditions. AMB Expr. 2019;9:5. doi: 10.1186/s13568-018-0727-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Katla S, Yoganand KNR, Hingane S, et al. Novel glycosylated human interferon alpha 2b expressed in glycoengineered Pichia pastoris and its biological activity: N-linked glycoengineering approach. Enzyme Microb Technol. 2019;128:49–58. doi: 10.1016/j.enzmictec.2019.05.007. [DOI] [PubMed] [Google Scholar]
  150. Kaye Y, Grundman O, Leu S, et al. Metabolic engineering toward enhanced LC-PUFA biosynthesis in Nannochloropsis oceanica : overexpression of endogenous Δ12 desaturase driven by stress-inducible promoter leads to enhanced deposition of polyunsaturated fatty acids in TAG. Algal Res. 2015;11:387–398. doi: 10.1016/j.algal.2015.05.003. [DOI] [Google Scholar]
  151. Kelley B (2009) Industrialization of mAb production technology: the bioprocessing industry at a crossroads. mAbs 1:443–452. 10.4161/mabs.1.5.9448 [DOI] [PMC free article] [PubMed]
  152. Kesavadev J, Murthy LS, Chaudhury T, et al. One-year safety and effectiveness of insulin degludec in patients with diabetes mellitus in routine clinical practice in India—TRUST (Tresiba real-world use study) Metabol Open. 2022;14:100184. doi: 10.1016/j.metop.2022.100184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Khavari F, Saidijam M, Taheri M, Nouri F. Microalgae: therapeutic potentials and applications. Mol Biol Rep. 2021;48:4757–4765. doi: 10.1007/s11033-021-06422-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Kiefer AM, Niemeyer J, Probst A, et al. Production and secretion of functional SARS-CoV-2 spike protein in Chlamydomonas reinhardtii. Front Plant Sci. 2022;13:988870. doi: 10.3389/fpls.2022.988870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Kim H, Kim J-S. A guide to genome engineering with programmable nucleases. Nat Rev Genet. 2014;15:321–334. doi: 10.1038/nrg3686. [DOI] [PubMed] [Google Scholar]
  156. Kim HJ, Kim H-J. Yeast as an expression system for producing virus-like particles: what factors do we need to consider? Lett Appl Microbiol. 2017;64:111–123. doi: 10.1111/lam.12695. [DOI] [PubMed] [Google Scholar]
  157. Kim SH, Lee GM. Down-regulation of lactate dehydrogenase-A by siRNAs for reduced lactic acid formation of Chinese hamster ovary cells producing thrombopoietin. Appl Microbiol Biotechnol. 2007;74:152–159. doi: 10.1007/s00253-006-0654-5. [DOI] [PubMed] [Google Scholar]
  158. Kim SH, Lee GM. Functional expression of human pyruvate carboxylase for reduced lactic acid formation of Chinese hamster ovary cells (DG44) Appl Microbiol Biotechnol. 2007;76:659–665. doi: 10.1007/s00253-007-1041-6. [DOI] [PubMed] [Google Scholar]
  159. Kim H, Yoo SJ, Kang HA. Yeast synthetic biology for the production of recombinant therapeutic proteins. FEMS Yeast Res. 2014 doi: 10.1111/1567-1364.12195. [DOI] [PubMed] [Google Scholar]
  160. Kim J, Lee S, Baek K, Jin E. Site-specific gene knock-out and on-site heterologous gene overexpression in Chlamydomonas reinhardtii via a CRISPR-Cas9-mediated knock-in method. Front Plant Sci. 2020;11:306. doi: 10.3389/fpls.2020.00306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Kimia Z, Hosseini SN, Ashraf Talesh SS, et al. A novel application of ion exchange chromatography in recombinant hepatitis B vaccine downstream processing: improving recombinant HBsAg homogeneity by removing associated aggregates. J Chromatogr B. 2019;1113:20–29. doi: 10.1016/j.jchromb.2019.03.009. [DOI] [PubMed] [Google Scholar]
  162. King EM, Mazor R, Çuburu N, Pastan I. Low-dose methotrexate prevents primary and secondary humoral immune responses and induces immune tolerance to a recombinant immunotoxin. J Immunol. 2018;200:2038–2045. doi: 10.4049/jimmunol.1701430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Kintzing JR, Filsinger Interrante MV, Cochran JR. Emerging strategies for developing next-generation protein therapeutics for cancer treatment. Trends Pharmacol Sci. 2016;37:993–1008. doi: 10.1016/j.tips.2016.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Kishimoto TK, Ferrari JD, LaMothe RA, et al. Improving the efficacy and safety of biologic drugs with tolerogenic nanoparticles. Nat Nanotechnol. 2016;11:890–899. doi: 10.1038/nnano.2016.135. [DOI] [PubMed] [Google Scholar]
  165. Klein CA, Emde L, Kuijpers A, Sobetzko P. MoCloFlex: a modular yet flexible cloning system. Front Bioeng Biotechnol. 2019;7:271. doi: 10.3389/fbioe.2019.00271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Kol S, Ley D, Wulff T, et al. Multiplex secretome engineering enhances recombinant protein production and purity. Nat Commun. 2020;11:1908. doi: 10.1038/s41467-020-15866-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Kontermann RE. Strategies for extended serum half-life of protein therapeutics. Curr Opin Biotechnol. 2011;22:868–876. doi: 10.1016/j.copbio.2011.06.012. [DOI] [PubMed] [Google Scholar]
  168. Kost TA, Kemp CW. Fundamentals of baculovirus expression and applications. In: Vega MC, editor. Advanced technologies for protein complex production and characterization. Cham: Springer International Publishing; 2016. pp. 187–197. [Google Scholar]
  169. Kota RK, Ambati RR, YVV AK, et al. Recent advances in probiotics as live biotherapeutics against gastrointestinal diseases. CPD. 2018;24:3162–3171. doi: 10.2174/1381612824666180717105128. [DOI] [PubMed] [Google Scholar]
  170. Krainer FW, Dietzsch C, Hajek T, et al. Recombinant protein expression in Pichia pastoris strains with an engineered methanol utilization pathway. Microb Cell Fact. 2012;11:22. doi: 10.1186/1475-2859-11-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Kumar D, Batra J, Komives C, Rathore A (2019a) QbD based media development for the production of fab fragments in E. coli. Bioengineering 6:29. 10.3390/bioengineering6020029 [DOI] [PMC free article] [PubMed]
  172. Kumar D, Batra J, Komives C, Rathore A (2019b) QbD based media development for the production of fab fragments in E. coli. Bioengineering 6:29. 10.3390/bioengineering6020029 [DOI] [PMC free article] [PubMed]
  173. Łącki KM, Riske FJ. Affinity chromatography: an enabling technology for large-scale bioprocessing. Biotechnol J. 2020;15:1800397. doi: 10.1002/biot.201800397. [DOI] [PubMed] [Google Scholar]
  174. Lagassé HAD, Alexaki A, Simhadri VL, et al (2017) Recent advances in (therapeutic protein) drug development. F1000Res 6:113. 10.12688/f1000research.9970.1 [DOI] [PMC free article] [PubMed]
  175. Lalonde M-E, Durocher Y. Therapeutic glycoprotein production in mammalian cells. J Biotechnol. 2017;251:128–140. doi: 10.1016/j.jbiotec.2017.04.028. [DOI] [PubMed] [Google Scholar]
  176. Larrick JW, Yu L, Naftzger C, et al. Production of secretory IgA antibodies in plants. Biomol Eng. 2001;18:87–94. doi: 10.1016/S1389-0344(01)00102-2. [DOI] [PubMed] [Google Scholar]
  177. Leader B, Baca QJ, Golan DE. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov. 2008;7:21–39. doi: 10.1038/nrd2399. [DOI] [PubMed] [Google Scholar]
  178. Lee JS, Grav LM, Lewis NE, Faustrup Kildegaard H. CRISPR/Cas9-mediated genome engineering of CHO cell factories: application and perspectives. Biotechnol J. 2015;10:979–994. doi: 10.1002/biot.201500082. [DOI] [PubMed] [Google Scholar]
  179. Lee AC-L, Harris JL, Khanna KK, Hong J-H. A Comprehensive review on current advances in peptide drug development and design. Int J Mol Sci. 2019;20:2383. doi: 10.3390/ijms20102383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Legastelois I, Buffin S, Peubez I, et al. Non-conventional expression systems for the production of vaccine proteins and immunotherapeutic molecules. Hum Vaccin Immunother. 2017;13:947–961. doi: 10.1080/21645515.2016.1260795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Levin D, Golding B, Strome SE, Sauna ZE. Fc fusion as a platform technology: potential for modulating immunogenicity. Trends Biotechnol. 2015;33:27–34. doi: 10.1016/j.tibtech.2014.11.001. [DOI] [PubMed] [Google Scholar]
  182. Levin P, Zhou S, Durden E, et al. Clinical and economic outcomes associated with the timing of initiation of basal insulin in patients with type 2 diabetes mellitus previously treated with oral antidiabetes drugs. Clin Ther. 2016;38:110–121. doi: 10.1016/j.clinthera.2015.11.011. [DOI] [PubMed] [Google Scholar]
  183. Li SW, Yu B, Byrne G, et al. Identification and CRISPR/Cas9 inactivation of the C1s protease responsible for proteolysis of recombinant proteins produced in CHO cells. Biotechnol Bioeng. 2019;116:2130–2145. doi: 10.1002/bit.27016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Li SW, Wright M, Healey JF, et al. Gene editing in CHO cells to prevent proteolysis and enhance glycosylation: production of HIV envelope proteins as vaccine immunogens. PLoS ONE. 2020;15:e0233866. doi: 10.1371/journal.pone.0233866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Lin AY, Dinner SN. Moxetumomab pasudotox for hairy cell leukemia: preclinical development to FDA approval. Blood Adv. 2019;3:2905–2910. doi: 10.1182/bloodadvances.2019000507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Lobstein J, Emrich CA, Jeans C, et al. SHuffle, a novel Escherichia coli protein expression strain capable of correctly folding disulfide bonded proteins in its cytoplasm. Microb Cell Fact. 2012;11:753. doi: 10.1186/1475-2859-11-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Loh H-S, Green BJ, Yusibov V. Using transgenic plants and modified plant viruses for the development of treatments for human diseases. Curr Opin Virol. 2017;26:81–89. doi: 10.1016/j.coviro.2017.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Lomonossoff GP, D’Aoust M-A. Plant-produced biopharmaceuticals: a case of technical developments driving clinical deployment. Science. 2016;353:1237–1240. doi: 10.1126/science.aaf6638. [DOI] [PubMed] [Google Scholar]
  189. Looser V, Bruhlmann B, Bumbak F, et al. Cultivation strategies to enhance productivity of Pichia pastoris: a review. Biotechnol Adv. 2015;33:1177–1193. doi: 10.1016/j.biotechadv.2015.05.008. [DOI] [PubMed] [Google Scholar]
  190. Love KR, Dalvie NC, Love JC. The yeast stands alone: the future of protein biologic production. Curr Opin Biotechnol. 2018;53:50–58. doi: 10.1016/j.copbio.2017.12.010. [DOI] [PubMed] [Google Scholar]
  191. Lozano Terol G, Gallego-Jara J, Sola Martínez RA, et al (2019) Engineering protein production by rationally choosing a carbon and nitrogen source using E. coli BL21 acetate metabolism knockout strains. Microb Cell Fact 18:151. 10.1186/s12934-019-1202-1 [DOI] [PMC free article] [PubMed]
  192. Luo B, Lee AS. The critical roles of endoplasmic reticulum chaperones and unfolded protein response in tumorigenesis and anticancer therapies. Oncogene. 2013;32:805–818. doi: 10.1038/onc.2012.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. MacKenzie IZ, Bichler J, Mason GC, et al. Efficacy and safety of a new, chromatographically purified rhesus (D) immunoglobulin. Eur J Obstetr Gynecol Reprod Biol. 2004;117:154–161. doi: 10.1016/j.ejogrb.2004.03.009. [DOI] [PubMed] [Google Scholar]
  194. Macri C, Dumont C, Johnston AP, Mintern JD. Targeting dendritic cells: a promising strategy to improve vaccine effectiveness. Clin Transl Immunology. 2016;5:e66. doi: 10.1038/cti.2016.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Madhavan A, Pandey A, Sukumaran RK. Expression system for heterologous protein expression in the filamentous fungus Aspergillus unguis. Biores Technol. 2017;245:1334–1342. doi: 10.1016/j.biortech.2017.05.140. [DOI] [PubMed] [Google Scholar]
  196. Maksimenko OG, Deykin AV, Khodarovich YM, Georgiev PG (2013) Use of transgenic animals in biotechnology: prospects and problems. Acta Naturae 5:33–46. 10.32607/20758251-2013-5-1-33-46 [PMC free article] [PubMed]
  197. Maksum IP, Nabiel A, Ishmayana S, Soedjanaatmadja UM. E. Coli Infections-importance of early diagnosis and efficient treatment. IntechOpen; 2020. Safety aspect of recombinant protein produced by Escherichia coli: toxin evaluation with strain and genomic approach. [Google Scholar]
  198. Manna S, Di Natale C, Florio D, Marasco D. Peptides as therapeutic agents for inflammatory-related diseases. Int J Mol Sci. 2018;19:2714. doi: 10.3390/ijms19092714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Manuell AL, Beligni MV, Elder JH, et al. Robust expression of a bioactive mammalian protein in Chlamydomonas chloroplast. Plant Biotechnol J. 2007;5:402–412. doi: 10.1111/j.1467-7652.2007.00249.x. [DOI] [PubMed] [Google Scholar]
  200. Martemyanov KA, Shirokov VA, Kurnasov OV, et al. Cell-free production of biologically active polypeptides: application to the synthesis of antibacterial peptide cecropin. Protein Expr Purif. 2001;21:456–461. doi: 10.1006/prep.2001.1400. [DOI] [PubMed] [Google Scholar]
  201. Marx V. Watching peptide drugs grow up. Chem Eng News. 2005;83:17–24. doi: 10.1021/cen-v083n011.p017. [DOI] [Google Scholar]
  202. Masuda Y, Tsuda M, Hashikawa‐Muto C, et al (2019) Cation exchange chromatography performed in overloaded mode is effective in removing viruses during the manufacturing of monoclonal antibodies. Biotechnol Prog. 10.1002/btpr.2858 [DOI] [PubMed]
  203. Mattanovich D, Branduardi P, Dato L, et al. Recombinant protein production in yeasts. In: Lorence A, et al., editors. Recombinant gene expression. Totowa: Humana Press; 2012. pp. 329–358. [DOI] [PubMed] [Google Scholar]
  204. Matthews CB, Wright C, Kuo A, et al. Reexamining opportunities for therapeutic protein production in eukaryotic microorganisms. Biotechnol Bioeng. 2017;114:2432–2444. doi: 10.1002/bit.26378. [DOI] [PubMed] [Google Scholar]
  205. Matucci A, Nencini F, Maggi E, Vultaggio A. Hypersensitivity reactions to biologics used in rheumatology. Expert Rev Clin Immunol. 2019;15:1263–1271. doi: 10.1080/1744666X.2020.1684264. [DOI] [PubMed] [Google Scholar]
  206. Mayfield SP, Franklin SE, Lerner RA. Expression and assembly of a fully active antibody in algae. Proc Natl Acad Sci USA. 2003;100:438–442. doi: 10.1073/pnas.0237108100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Mazor R, King EM, Onda M, et al. Tolerogenic nanoparticles restore the antitumor activity of recombinant immunotoxins by mitigating immunogenicity. Proc Natl Acad Sci USA. 2018 doi: 10.1073/pnas.1717063115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. McCarthy DP, Yap JW-T, Harp CT, et al. An antigen-encapsulating nanoparticle platform for TH1/17 immune tolerance therapy. Nanomedicine. 2017;13:191–200. doi: 10.1016/j.nano.2016.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. McLaughlin M, Vandenbroeck K. The endoplasmic reticulum protein folding factory and its chaperones: new targets for drug discovery?: ER chaperones as targets in drug discovery. Br J Pharmacol. 2011;162:328–345. doi: 10.1111/j.1476-5381.2010.01064.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Meade E, Hehir S, Rowan N, Garvey M. Mycotherapy: potential of fungal bioactives for the treatment of mental health disorders and morbidities of chronic pain. JoF. 2022;8:290. doi: 10.3390/jof8030290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Mengdai X, Xiaoxing M, Hao H, et al. Yeast beta-Glucan effects on gut microbiota for regulating insulin signaling system on alzheimer’s disease. Proc Nutr Soc. 2020;79(OCE2):E263. doi: 10.1017/s0029665120002116. [DOI] [Google Scholar]
  212. Merritt JH, Ollis AA, Fisher AC, DeLisa MP. Glycans-by-design: engineering bacteria for the biosynthesis of complex glycans and glycoconjugates. Biotechnol Bioeng. 2013;110:1550–1564. doi: 10.1002/bit.24885. [DOI] [PubMed] [Google Scholar]
  213. Mf P, Va T, Sj C, et al (2021) Single-use systems bioreactors in the biopharmaceutical industry and its use in SARS-CoV-2 candidate vaccine production—a review. Prensa Med Argent. 10.47275/0032-745X-348
  214. Miceli M, Cutignano A, Conte M, et al. Monoacylglycerides from the Diatom Skeletonema marinoi induce selective cell death in cancer cells. Mar Drugs. 2019;17:625. doi: 10.3390/md17110625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Miguel S, Nisse E, Biteau F, et al. Assessing carnivorous plants for the production of recombinant proteins. Front Plant Sci. 2019;10:793. doi: 10.3389/fpls.2019.00793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Monteil V, Kwon H, Prado P, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181:905–913.e7. doi: 10.1016/j.cell.2020.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Monzani PS, Adona PR, Ohashi OM, et al. Transgenic bovine as bioreactors: challenges and perspectives. Bioengineered. 2016;7:123–131. doi: 10.1080/21655979.2016.1171429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Moura RR, Melo LM, Freitas VJDF. Production of recombinant proteins in milk of transgenic and non-transgenic goats. Braz Arch Biol Technol. 2011;54:927–938. doi: 10.1590/S1516-89132011000500010. [DOI] [Google Scholar]
  219. Moustafa K, Makhzoum A, Trémouillaux-Guiller J. Molecular farming on rescue of pharma industry for next generations. Crit Rev Biotechnol. 2016;36:840–850. doi: 10.3109/07388551.2015.1049934. [DOI] [PubMed] [Google Scholar]
  220. Moyade P, Vinjamuri S. The heart matters: a review of incidental cardiac uptake on Ga-68 DOTA peptide PET-CT scans. Nucl Med Commun. 2019;40:1081–1085. doi: 10.1097/MNM.0000000000001064. [DOI] [PubMed] [Google Scholar]
  221. Murashima K, Chen C-L, Kosugi A, et al. Heterologous production of clostridium cellulovorans engB, using protease-deficient bacillus subtilis, and preparation of active recombinant cellulosomes. J Bacteriol. 2002;184:76–81. doi: 10.1128/JB.184.1.76-81.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Nakar C, Shapiro A. Hemophilia A with inhibitor: immune tolerance induction (ITI) in the mirror of time. Transfus Apher Sci. 2019;58:578–589. doi: 10.1016/j.transci.2019.08.008. [DOI] [PubMed] [Google Scholar]
  223. Narayanan H, Sokolov M, Butté A, Morbidelli M (2019) Decision Tree‐PLS (DT‐PLS) algorithm for the development of process: Specific local prediction models. Biotechnol Progr. 10.1002/btpr.2818 [DOI] [PubMed]
  224. Naz Z, Kanwal A, Tayyeb A. A Review on empirical approach to therapeutic recombinant protein production factories: applications, pharmacokinetics and challenges. J Bioresour Manag. 2022;9:16. [Google Scholar]
  225. Neville JJ, Orlando J, Mann K, et al. Ubiquitous chromatin-opening elements (UCOEs): applications in biomanufacturing and gene therapy. Biotechnol Adv. 2017;35:557–564. doi: 10.1016/j.biotechadv.2017.05.004. [DOI] [PubMed] [Google Scholar]
  226. Nguyen VD, Hatahet F, Salo KE, et al (2011) Pre-expression of a sulfhydryl oxidase significantly increases the yields of eukaryotic disulfide bond containing proteins expressed in the cytoplasm of E.coli. Microb Cell Fact 10:1. 10.1186/1475-2859-10-1 [DOI] [PMC free article] [PubMed]
  227. Nielsen J. Production of biopharmaceutical proteins by yeast: advances through metabolic engineering. Bioengineered. 2013;4:207–211. doi: 10.4161/bioe.22856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Nishida T, Kubota S, Takigawa M. Production of recombinant CCN2 protein by mammalian cells. In: Takigawa M, editor. CCN Proteins. New York: Springer; 2017. pp. 95–105. [DOI] [PubMed] [Google Scholar]
  229. Nunberg JH, Meade JH, Cole G, et al. Molecular cloning and characterization of the glucoamylase gene of Aspergillus awamori. Mol Cell Biol. 1984;4:2306–2315. doi: 10.1128/mcb.4.11.2306-2315.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Oates JA, Wood AJJ, Jankovic J, Brin MF. Therapeutic uses of botulinum toxin. N Engl J Med. 1991;324:1186–1194. doi: 10.1056/NEJM199104253241707. [DOI] [PubMed] [Google Scholar]
  231. Oeller M, Kang RJD, Bolt HL, et al. Sequence-based prediction of the intrinsic solubility of peptides containing non-natural amino acids. Nat Commun. 2023;14:7475. doi: 10.1038/s41467-023-42940-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Ohya S, Matsuda T. Poly (N-isopropylacrylamide) (PNIPAM)-grafted gelatin as thermoresponsive three-dimensional artificial extracellular matrix: molecular and formulation parameters vs. cell proliferation potential. J Biomater Sci Polym Ed. 2005;16:809–827. doi: 10.1163/1568562054255736. [DOI] [PubMed] [Google Scholar]
  233. Osz-Papai J, Radu L, Abdulrahman W, et al. Insect cells-baculovirus system for the production of difficult to express proteins. In: García-Fruitós E, et al., editors. Insoluble proteins. New York: Springer; 2015. pp. 181–205. [DOI] [PubMed] [Google Scholar]
  234. Owczarek B, Gerszberg A, Hnatuszko-Konka K. A brief reminder of systems of production and chromatography-based recovery of recombinant protein biopharmaceuticals. Biomed Res Int. 2019;2019:1–13. doi: 10.1155/2019/4216060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Padhi A, Sengupta M, Sengupta S, et al. Antimicrobial peptides and proteins in mycobacterial therapy: current status and future prospects. Tuberculosis. 2014;94:363–373. doi: 10.1016/j.tube.2014.03.011. [DOI] [PubMed] [Google Scholar]
  236. Pandhal J, Wright PC. N-Linked glycoengineering for human therapeutic proteins in bacteria. Biotechnol Lett. 2010;32:1189–1198. doi: 10.1007/s10529-010-0289-6. [DOI] [PubMed] [Google Scholar]
  237. Papaneophytou CP, Kontopidis GA. Optimization of TNF-α overexpression in Escherichia coli using response surface methodology: purification of the protein and oligomerization studies. Protein Expr Purif. 2012;86:35–44. doi: 10.1016/j.pep.2012.09.002. [DOI] [PubMed] [Google Scholar]
  238. Paraskevopoulou V, Falcone F. Polyionic tags as enhancers of protein solubility in recombinant protein expression. Microorganisms. 2018;6:47. doi: 10.3390/microorganisms6020047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Park KY, Wi SJ. Potential of plants to produce recombinant protein products. J Plant Biol. 2016;59:559–568. doi: 10.1007/s12374-016-0482-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Parsaie Nasab F, Aebi M, Bernhard G, Frey AD. A combined system for engineering glycosylation efficiency and glycan structure in saccharomyces cerevisiae. Appl Environ Microbiol. 2013;79:997–1007. doi: 10.1128/AEM.02817-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Pastan I, Hassan R, FitzGerald DJ, Kreitman RJ. Immunotoxin therapy of cancer. Nat Rev Cancer. 2006;6:559–565. doi: 10.1038/nrc1891. [DOI] [PubMed] [Google Scholar]
  242. Patel SG, Sayers EJ, He L, et al. Cell-penetrating peptide sequence and modification dependent uptake and subcellular distribution of green florescent protein in different cell lines. Sci Rep. 2019;9:6298. doi: 10.1038/s41598-019-42456-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Pathak M, Dutta D, ProfA R. Analytical QbD: development of a native gel electrophoresis method for measurement of monoclonal antibody aggregates. ELECTROPHORESIS. 2014 doi: 10.1002/elps.201400055. [DOI] [PubMed] [Google Scholar]
  244. Pato TP, Souza MCO, Mattos DA, et al. Purification of yellow fever virus produced in Vero cells for inactivated vaccine manufacture. Vaccine. 2019;37:3214–3220. doi: 10.1016/j.vaccine.2019.04.077. [DOI] [PubMed] [Google Scholar]
  245. Pearson S, Kietsiriroje N, Ajjan RA. Oral semaglutide in the management of type 2 diabetes: a report on the evidence to date. Diabetes Metab Syndr Obes. 2019;12:2515–2529. doi: 10.2147/DMSO.S229802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Penna G, Amuchastegui S, Laverny G, Adorini L. Vitamin D receptor agonists in the treatment of autoimmune diseases: selective targeting of myeloid but not plasmacytoid dendritic cells. J Bone Miner Res. 2007;22(Suppl 2):V69–73. doi: 10.1359/jbmr.07s217. [DOI] [PubMed] [Google Scholar]
  247. Pero J, Sloma A (1993) Proteases. In: Bacillus subtilis and other gram‐positive bacteria: biochemistry, physiology, and molecular genetics, pp 939–952. 10.1128/9781555818388.ch63
  248. Pieters R, Hunger SP, Boos J, et al. L-asparaginase treatment in acute lymphoblastic leukemia: a focus on Erwinia asparaginase. Cancer. 2011;117:238–249. doi: 10.1002/cncr.25489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Polez S, Origi D, Zahariev S, et al. A simplified and efficient process for insulin production in Pichia pastoris. PLoS ONE. 2016;11:e0167207. doi: 10.1371/journal.pone.0167207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Potla Durthi C, Pola M, Kola AK, Rajulapati SB (2019) Screening, optimization of culture conditions and scale-up for production of the L-Glutaminase by novel isolated Bacillus sps. mutant endophyte using response surface methodology. Biocatal Agric Biotechnol 18:101077. 10.1016/j.bcab.2019.101077
  251. Qarawi MA, Farah FH, Mohamed SS, El Haj BM. A review on therapeutic proteins: stability aspects and clinical implications. Int Res J Pharm. 2019;10:1–7. doi: 10.7897/2230-8407.100233. [DOI] [Google Scholar]
  252. Raab N, Mathias S, Alt K, et al. CRISPR/Cas9-mediated knockout of microRNA-744 improves antibody titer of CHO production cell lines. Biotechnol J. 2019;14:1800477. doi: 10.1002/biot.201800477. [DOI] [PubMed] [Google Scholar]
  253. Rahimi A, Hosseini SN, Karimi A, et al. Enhancing the efficiency of recombinant hepatitis B surface antigen production in Pichia pastoris by employing continuous fermentation. Biochem Eng J. 2019;141:112–119. doi: 10.1016/j.bej.2018.10.019. [DOI] [Google Scholar]
  254. Rajendra Y, Balasubramanian S, Peery RB, et al. Bioreactor scale up and protein product quality characterization of piggyBac transposon derived CHO pools. Biotechnol Progress. 2017;33:534–540. doi: 10.1002/btpr.2447. [DOI] [PubMed] [Google Scholar]
  255. Ramos-de-la-Peña AM, González-Valdez J, Aguilar O. Protein A chromatography: challenges and progress in the purification of monoclonal antibodies. J Sep Sci. 2019;42:1816–1827. doi: 10.1002/jssc.201800963. [DOI] [PubMed] [Google Scholar]
  256. Rao TSS, Andrade C. Bremelanotide for hypoactive sexual desire disorder. J Psychosex Health. 2020;2:13–15. doi: 10.1177/2631831820909450. [DOI] [Google Scholar]
  257. Rasala BA, Mayfield SP. Photosynthetic biomanufacturing in green algae; production of recombinant proteins for industrial, nutritional, and medical uses. Photosynth Res. 2015;123:227–239. doi: 10.1007/s11120-014-9994-7. [DOI] [PubMed] [Google Scholar]
  258. Reddy PH, Johnson AMA, Kumar JK, et al. Heterologous expression of Infectious bursal disease virus VP2 gene in Chlorella pyrenoidosa as a model system for molecular farming. Plant Cell Tiss Organ Cult. 2017;131:119–126. doi: 10.1007/s11240-017-1268-6. [DOI] [Google Scholar]
  259. Reinhart D, Damjanovic L, Kaisermayer C, et al. Bioprocessing of recombinant CHO-K1, CHO-DG44, and CHO-S: CHO expression hosts favor either mAb production or biomass synthesis. Biotechnol J. 2019;14:1700686. doi: 10.1002/biot.201700686. [DOI] [PubMed] [Google Scholar]
  260. Reyes SJ, Durocher Y, Pham PL, Henry O. Modern sensor tools and techniques for monitoring, controlling, and improving cell culture processes. Processes. 2022;10:189. doi: 10.3390/pr10020189. [DOI] [Google Scholar]
  261. Rita Costa A, Elisa Rodrigues M, Henriques M, et al. Guidelines to cell engineering for monoclonal antibody production. Eur J Pharm Biopharm. 2010;74:127–138. doi: 10.1016/j.ejpb.2009.10.002. [DOI] [PubMed] [Google Scholar]
  262. Rizwan M, Mujtaba G, Memon SA, et al. Exploring the potential of microalgae for new biotechnology applications and beyond: a review. Renew Sustain Energy Rev. 2018;92:394–404. doi: 10.1016/j.rser.2018.04.034. [DOI] [Google Scholar]
  263. Robinson SD, Safavi-Hemami H, McIntosh LD, et al. Diversity of conotoxin gene superfamilies in the venomous snail, conus victoriae. PLOS ONE. 2014;9:e87648. doi: 10.1371/journal.pone.0087648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Román R, Miret J, Roura A, et al. Enabling HEK293 cells for antibiotic-free media bioprocessing through CRISPR/Cas9 gene editing. Biochem Eng J. 2019;151:107299. doi: 10.1016/j.bej.2019.107299. [DOI] [Google Scholar]
  265. Roth DA, Tawa NE, O’Brien JM, et al. Nonviral transfer of the gene encoding coagulation factor VIII in patients with severe hemophilia A. N Engl J Med. 2001;344:1735–1742. doi: 10.1056/NEJM200106073442301. [DOI] [PubMed] [Google Scholar]
  266. Rozov SM, Deineko EV. Strategies for optimizing recombinant protein synthesis in plant cells: classical approaches and new directions. Mol Biol. 2019;53:157–175. doi: 10.1134/S0026893319020146. [DOI] [PubMed] [Google Scholar]
  267. Rozov SM, Permyakova NV, Deineko EV. Main strategies of plant expression system glycoengineering for producing humanized recombinant pharmaceutical proteins. Biochemistry Moscow. 2018;83:215–232. doi: 10.1134/S0006297918030033. [DOI] [PubMed] [Google Scholar]
  268. Rutgers T, Gordon D, Gathoye AM, et al. Hepatitis B surface antigen as carrier matrix for the repetitive epitope of the circumsporozoite protein of plasmodium falciparum. Nat Biotechnol. 1988;6:1065–1070. doi: 10.1038/nbt0988-1065. [DOI] [Google Scholar]
  269. Sagiya Y, Yamagata H, Udaka S. Direct high-level secretion into the culture medium of tuna growth hormone in biologically active form by Bacillus brevis. Appl Microbiol Biotechnol. 1994;42:358–363. doi: 10.1007/BF00902742. [DOI] [PubMed] [Google Scholar]
  270. Sakuma T, Takenaga M, Kawabe Y, et al. Homologous recombination-independent large gene cassette knock-in in CHO cells using TALEN and MMEJ-directed donor plasmids. IJMS. 2015;16:23849–23866. doi: 10.3390/ijms161023849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Salazar-Fontana LI, Desai DD, Khan TA, et al. Approaches to mitigate the unwanted immunogenicity of therapeutic proteins during drug development. AAPS J. 2017;19:377–385. doi: 10.1208/s12248-016-0030-z. [DOI] [PubMed] [Google Scholar]
  272. Saltz LB, Meropol NJ, Loehrer PJ, et al. Phase II trial of cetuximab in patients with refractory colorectal cancer that expresses the epidermal growth factor receptor. JCO. 2004;22:1201–1208. doi: 10.1200/JCO.2004.10.182. [DOI] [PubMed] [Google Scholar]
  273. Saraswat M, Musante L, Ravidá A, et al. Preparative purification of recombinant proteins: current status and future trends. Biomed Res Int. 2013;2013:1–18. doi: 10.1155/2013/312709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Saraya R, Gidijala L, Veenhuis M, Van Der Klei IJ. Tools for genetic engineering of the yeast hansenula polymorpha. In: Mapelli V, editor. Yeast Metabolic Engineering. New York, New York, NY: Springer; 2014. pp. 43–62. [DOI] [PubMed] [Google Scholar]
  275. Sato D, Nozaki T. Methionine gamma-lyase: the unique reaction mechanism, physiological roles, and therapeutic applications against infectious diseases and cancers. IUBMB Life. 2009;61:1019–1028. doi: 10.1002/iub.255. [DOI] [PubMed] [Google Scholar]
  276. Saul D, Sudbery P. Molecular cloning of WHI2, a gene involved in the regulation of cell proliferation in Saccharomyces cerevisiae. Microbiology. 1985;131:1797–1806. doi: 10.1099/00221287-131-7-1797. [DOI] [PubMed] [Google Scholar]
  277. Sauna ZE, Lagassé D, Pedras-Vasconcelos J, et al. Evaluating and mitigating the immunogenicity of therapeutic proteins. Trends Biotechnol. 2018;36:1068–1084. doi: 10.1016/j.tibtech.2018.05.008. [DOI] [PubMed] [Google Scholar]
  278. Sawatzki A, Hans S, Narayanan H, et al. Accelerated bioprocess development of endopolygalacturonase-production with saccharomyces cerevisiae using multivariate prediction in a 48 mini-bioreactor automated platform. Bioengineering. 2018;5:101. doi: 10.3390/bioengineering5040101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Schillberg S, Raven N, Spiegel H, et al. Critical analysis of the commercial potential of plants for the production of recombinant proteins. Front Plant Sci. 2019;10:720. doi: 10.3389/fpls.2019.00720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Schmidt SR. Process intensification based on disposable solutions as first step toward continuous processing. In: Subramanian G, editor. Process control, intensification, and digitalisation in continuous biomanufacturing. 1. Wiley; 2022. pp. 137–178. [Google Scholar]
  281. Schmuck B, Chen G, Pelcman J, et al (2021) Expression of the human molecular chaperone domain Bri2 BRICHOS on a gram per liter scale with an E. coli fed-batch culture. Microb Cell Fact 20:150. 10.1186/s12934-021-01638-8 [DOI] [PMC free article] [PubMed]
  282. Seo H-S, Park J-S, Han K-Y, et al. Analysis and characterization of hepatitis B vaccine particles synthesized from Hansenula polymorpha. Vaccine. 2008;26:4138–4144. doi: 10.1016/j.vaccine.2008.05.070. [DOI] [PubMed] [Google Scholar]
  283. Seyis I, Aksoz N. Effect of carbon and nitrogen sources on xylanase production by Trichoderma harzianum 1073 D3. Int Biodeterior Biodegrad. 2005;55:115–119. doi: 10.1016/j.ibiod.2004.09.001. [DOI] [Google Scholar]
  284. Shaikh R, O’Brien DP, Croker DM, Walker GM (2018) The development of a pharmaceutical oral solid dosage forms. In: Computer aided chemical engineering. Elsevier, New York, pp 27–65
  285. Shankar R, Upadhyay PK, Kumar M. Protease enzymes: highlights on potential of proteases as therapeutics agents. Int J Pept Res Ther. 2021;27:1281–1296. doi: 10.1007/s10989-021-10167-2. [DOI] [Google Scholar]
  286. Sharker SMd, Rahman A. A review on the current methods of chinese hamster ovary (CHO) cells cultivation for the production of therapeutic protein. CDDT. 2021;18:354–364. doi: 10.2174/1570163817666200312102137. [DOI] [PubMed] [Google Scholar]
  287. Sharma A, Herekar AA, Bhagatwala J, Rao SS. Profile of plecanatide in the treatment of chronic idiopathic constipation: design, development, and place in therapy. Clin Exp Gastroenterol. 2019;12:31–36. doi: 10.2147/CEG.S145668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Shekhawat LK, Godara A, Kumar V, Rathore AS. Design of experiments applications in bioprocessing: chromatography process development using split design of experiments. Biotechnol Prog. 2019;35:e2730. doi: 10.1002/btpr.2730. [DOI] [PubMed] [Google Scholar]
  289. Shepelev MV, Kalinichenko SV, Deykin AV, Korobko IV (2018) Production of recombinant proteins in the milk of transgenic animals: current state and prospects. Acta Naturae 10:40–47. 10.32607/20758251-2018-10-3-40-47 [PMC free article] [PubMed]
  290. Shih HH. Discovery process for antibody-based therapeutics. In: Tabrizi MA, Bornstein GG, Klakamp SL, editors. Development of antibody-based therapeutics. New York: Springer; 2012. pp. 9–32. [Google Scholar]
  291. Shrivastava A, Pal M, Sharma RK. Pichia as yeast cell factory for production of industrially important bio-products: Current trends, challenges, and future prospects. J Bioresour Bioprod. 2023;8:108–124. doi: 10.1016/j.jobab.2023.01.007. [DOI] [Google Scholar]
  292. Shukla AA, Thömmes J. Recent advances in large-scale production of monoclonal antibodies and related proteins. Trends Biotechnol. 2010;28:253–261. doi: 10.1016/j.tibtech.2010.02.001. [DOI] [PubMed] [Google Scholar]
  293. Slattery SS, Giguere DJ, Stuckless EE, et al. Phosphate-regulated expression of the SARS-CoV-2 receptor-binding domain in the diatom Phaeodactylum tricornutum for pandemic diagnostics. Sci Rep. 2022;12:7010. doi: 10.1038/s41598-022-11053-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Sodee DB, Malguria N, Faulhaber P, et al. Multicenter ProstaScint imaging findings in 2154 patients with prostate cancer. Urology. 2000;56:988–993. doi: 10.1016/S0090-4295(00)00824-4. [DOI] [PubMed] [Google Scholar]
  295. Solá RJ, Griebenow K. Glycosylation of therapeutic proteins: an effective strategy to optimize efficacy. BioDrugs. 2010;24:9–21. doi: 10.2165/11530550-000000000-00000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Soria-Guerra RE, Ramírez-Alonso JI, Ibáñez-Salazar A, et al. Expression of an HBcAg-based antigen carrying angiotensin II in Chlamydomonas reinhardtii as a candidate hypertension vaccine. Plant Cell Tiss Organ Cult. 2014;116:133–139. doi: 10.1007/s11240-013-0388-x. [DOI] [Google Scholar]
  297. Specht EA, Mayfield SP. Algae-based oral recombinant vaccines. Front Microbiol. 2014;5:76662. doi: 10.3389/fmicb.2014.00060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Sproles AE, Fields FJ, Smalley TN, et al. Recent advancements in the genetic engineering of microalgae. Algal Res. 2021;53:102158. doi: 10.1016/j.algal.2020.102158. [DOI] [Google Scholar]
  299. Stebbings R, Poole S, Thorpe R. Safety of biologics, lessons learnt from TGN1412. Curr Opin Biotechnol. 2009;20:673–677. doi: 10.1016/j.copbio.2009.10.002. [DOI] [PubMed] [Google Scholar]
  300. Svahn KS, Chryssanthou E, Olsen B, et al. Penicillium nalgiovense Laxa isolated from Antarctica is a new source of the antifungal metabolite amphotericin B. Fungal Biol Biotechnol. 2015;2:1. doi: 10.1186/s40694-014-0011-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Tai W, He L, Zhang X, et al. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell Mol Immunol. 2020;17:613–620. doi: 10.1038/s41423-020-0400-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Talukdar J, Dasgupta S, Nagle V, Bhadra B. COVID-19: potential of microalgae derived natural astaxanthin as adjunctive supplement in alleviating cytokine storm. SSRN J. 2020 doi: 10.2139/ssrn.3579738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Tang H, Wang S, Wang J, et al. N-hypermannose glycosylation disruption enhances recombinant protein production by regulating secretory pathway and cell wall integrity in Saccharomyces cerevisiae. Sci Rep. 2016;6:25654. doi: 10.1038/srep25654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Tekoah Y, Shulman A, Kizhner T, et al. Large-scale production of pharmaceutical proteins in plant cell culture-the protalix experience. Plant Biotechnol J. 2015;13:1199–1208. doi: 10.1111/pbi.12428. [DOI] [PubMed] [Google Scholar]
  305. Terpe K. Overview of bacterial expression systems for heterologous protein production: from molecular and biochemical fundamentals to commercial systems. Appl Microbiol Biotechnol. 2006;72:211–222. doi: 10.1007/s00253-006-0465-8. [DOI] [PubMed] [Google Scholar]
  306. Tesfaw A, Assefa F. Current trends in bioethanol production by Saccharomyces cerevisiae : substrate, inhibitor reduction, growth variables, coculture, and immobilization. Int Scholarly Res Notices. 2014;2014:1–11. doi: 10.1155/2014/532852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Tie Y, Tang F, Wei Y, Wei X. Immunosuppressive cells in cancer: mechanisms and potential therapeutic targets. J Hematol Oncol. 2022;15:61. doi: 10.1186/s13045-022-01282-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Tihanyi B, Nyitray L. Recent advances in CHO cell line development for recombinant protein production. Drug Discov Today Technol. 2020;38:25–34. doi: 10.1016/j.ddtec.2021.02.003. [DOI] [PubMed] [Google Scholar]
  309. Ton C, Stabile V, Carey E, et al. Development and scale-up of rVSV-SARS-CoV-2 vaccine process using single use bioreactor. Biotechnol Rep. 2023;37:e00782. doi: 10.1016/j.btre.2023.e00782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  310. Tong P, Hong Y, Xiao Y, et al. High production of laccase by a new basidiomycete, Trametes sp. Biotechnol Lett. 2007;29:295–301. doi: 10.1007/s10529-006-9241-1. [DOI] [PubMed] [Google Scholar]
  311. De La Torre BG, Albericio F (2020) Peptide therapeutics 2.0. Molecules 25:2293. 10.3390/molecules25102293 [DOI] [PMC free article] [PubMed]
  312. Tran M, Zhou B, Pettersson PL, et al. Synthesis and assembly of a full-length human monoclonal antibody in algal chloroplasts. Biotechnol Bioeng. 2009 doi: 10.1002/bit.22446. [DOI] [PubMed] [Google Scholar]
  313. Tran M, Van C, Barrera DJ, et al. Production of unique immunotoxin cancer therapeutics in algal chloroplasts. Proc Natl Acad Sci USA. 2013 doi: 10.1073/pnas.1214638110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Trassaert M, Vandermies M, Carly F, et al. New inducible promoter for gene expression and synthetic biology in Yarrowia lipolytica. Microb Cell Fact. 2017;16:141. doi: 10.1186/s12934-017-0755-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Tripathi NK, Shrivastava A. Recent developments in bioprocessing of recombinant proteins: expression hosts and process development. Front Bioeng Biotechnol. 2019;7:420. doi: 10.3389/fbioe.2019.00420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Tsomaia N. Peptide therapeutics: targeting the undruggable space. Eur J Med Chem. 2015;94:459–470. doi: 10.1016/j.ejmech.2015.01.014. [DOI] [PubMed] [Google Scholar]
  317. Turki S, Mrabet G, Jabloun Z, et al. A highly stable Yarrowia lipolytica lipase formulation for the treatment of pancreatic exocrine insufficiency. Biotechnol Appl Biochem. 2010;57:139–149. doi: 10.1042/BA20100272. [DOI] [PubMed] [Google Scholar]
  318. Udaka S, Yamagata H (2020) Extremely efficient protein secretion system in Bacillus brewis. Recombinant Microbes for Industrial and Agricultural Applications 71–79 [PubMed]
  319. Unni S, Prabhu AA, Pandey R, et al. Artificial neural network-genetic algorithm (ANN-GA) based medium optimization for the production of human interferon gamma (hIFN-γ) in Kluyveromyces lactis cell factory. Can J Chem Eng. 2019;97:843–858. doi: 10.1002/cjce.23350. [DOI] [Google Scholar]
  320. Utomo JC, Hodgins CL, Ro D-K. Multiplex genome editing in yeast by CRISPR/Cas9—a potent and agile tool to reconstruct complex metabolic pathways. Front Plant Sci. 2021;12:719148. doi: 10.3389/fpls.2021.719148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Valderrama-Rincon JD, Fisher AC, Merritt JH, et al. An engineered eukaryotic protein glycosylation pathway in Escherichia coli. Nat Chem Biol. 2012;8:434–436. doi: 10.1038/nchembio.921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Van Roy M, Ververken C, Beirnaert E, et al. The preclinical pharmacology of the high affinity anti-IL-6R Nanobody® ALX-0061 supports its clinical development in rheumatoid arthritis. Arthritis Res Ther. 2015;17:135. doi: 10.1186/s13075-015-0651-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. van Zonneveld M, Honkoop P, Hansen BE, et al. Long-term follow-up of alpha-interferon treatment of patients with chronic hepatitis B. Hepatology. 2004;39:804–810. doi: 10.1002/hep.20128. [DOI] [PubMed] [Google Scholar]
  324. Vandermies M, Fickers P. Bioreactor-scale strategies for the production of recombinant protein in the yeast Yarrowia lipolytica. Microorganisms. 2019;7:40. doi: 10.3390/microorganisms7020040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Vanier G, Hempel F, Chan P, et al. Biochemical Characterization of human anti-hepatitis B monoclonal antibody produced in the microalgae Phaeodactylum tricornutum. PLoS ONE. 2015;10:e0139282. doi: 10.1371/journal.pone.0139282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Varki A. Biological roles of glycans. Glycobiology. 2017;27:3–49. doi: 10.1093/glycob/cww086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Verhoef JJF, Carpenter JF, Anchordoquy TJ, Schellekens H. Potential induction of anti-PEG antibodies and complement activation toward PEGylated therapeutics. Drug Discovery Today. 2014;19:1945–1952. doi: 10.1016/j.drudis.2014.08.015. [DOI] [PubMed] [Google Scholar]
  328. Vieira Gomes A, Souza Carmo T, Silva Carvalho L, et al. Comparison of yeasts as hosts for recombinant protein production. Microorganisms. 2018;6:38. doi: 10.3390/microorganisms6020038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Walsh G. Biopharmaceutical benchmarks 2010. Nat Biotechnol. 2010;28:917–924. doi: 10.1038/nbt0910-917. [DOI] [PubMed] [Google Scholar]
  330. Walsh G. Biopharmaceutical benchmarks 2018. Nat Biotechnol. 2018;36:1136–1145. doi: 10.1038/nbt.4305. [DOI] [PubMed] [Google Scholar]
  331. Wang P, Sidney J, Dow C, et al. A systematic assessment of MHC class II peptide binding predictions and evaluation of a consensus approach. PLoS Comput Biol. 2008;4:e1000048. doi: 10.1371/journal.pcbi.1000048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Wang Y, Zhao S, Bai L, et al. Expression systems and species used for transgenic animal bioreactors. Biomed Res Int. 2013;2013:1–9. doi: 10.1155/2013/580463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Wang H, Chen X, Huang Z, et al. Expression and purification of porcine PID1 gene in Escherichia coli. Turk J Biol. 2014;38:523–527. doi: 10.3906/biy-1403-38. [DOI] [Google Scholar]
  334. Wang G, Huang M, Nielsen J. Exploring the potential of Saccharomyces cerevisiae for biopharmaceutical protein production. Curr Opin Biotechnol. 2017;48:77–84. doi: 10.1016/j.copbio.2017.03.017. [DOI] [PubMed] [Google Scholar]
  335. Wang R, Pan W, Jin L, et al. Human papillomavirus vaccine against cervical cancer: opportunity and challenge. Cancer Lett. 2020;471:88–102. doi: 10.1016/j.canlet.2019.11.039. [DOI] [PubMed] [Google Scholar]
  336. Wang L, Wang N, Zhang W, et al. Therapeutic peptides: current applications and future directions. Sig Transduct Target Ther. 2022;7:48. doi: 10.1038/s41392-022-00904-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Wannathong T, Waterhouse JC, Young REB, et al. New tools for chloroplast genetic engineering allow the synthesis of human growth hormone in the green alga Chlamydomonas reinhardtii. Appl Microbiol Biotechnol. 2016;100:5467–5477. doi: 10.1007/s00253-016-7354-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Ward M, Lin C, Victoria DC, et al. Characterization of humanized antibodies secreted by Aspergillus niger. Appl Environ Microbiol. 2004;70:2567–2576. doi: 10.1128/AEM.70.5.2567-2576.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Weesner JA, Annunziata I, Yang T, et al. Preclinical enzyme replacement therapy with a recombinant β-galactosidase-lectin fusion for CNS delivery and treatment of GM1-gangliosidosis. Cells. 2022;11:2579. doi: 10.3390/cells11162579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Weigel T, Soliman R, Wolff MW, Reichl U. Hydrophobic-interaction chromatography for purification of influenza A and B virus. J Chromatogr B. 2019;1117:103–117. doi: 10.1016/j.jchromb.2019.03.037. [DOI] [PubMed] [Google Scholar]
  341. Werten MWT, Eggink G, Cohen Stuart MA, de Wolf FA. Production of protein-based polymers in Pichia pastoris. Biotechnol Adv. 2019;37:642–666. doi: 10.1016/j.biotechadv.2019.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Wilken LR, Nikolov ZL. Recovery and purification of plant-made recombinant proteins. Biotechnol Adv. 2012;30:419–433. doi: 10.1016/j.biotechadv.2011.07.020. [DOI] [PubMed] [Google Scholar]
  343. Witmer C, Young G. Factor VIII inhibitors in hemophilia A: rationale and latest evidence. Ther Adv Hematol. 2013;4:59–72. doi: 10.1177/2040620712464509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  344. Witzig TE, Gordon LI, Cabanillas F, et al. Randomized controlled trial of yttrium-90–labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed b-cell non-hodgkin’s lymphoma. JCO. 2002;20:2453–2463. doi: 10.1200/JCO.2002.11.076. [DOI] [PubMed] [Google Scholar]
  345. Xiang Z-X, Gong J-S, Shi J-H, et al. High-efficiency secretory expression and characterization of the recombinant type III human-like collagen in Pichia pastoris. Bioresour Bioprocess. 2022;9:117. doi: 10.1186/s40643-022-00605-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Xu J, Towler M, Weathers PJ. Platforms for plant-based protein production. In: Pavlov A, Bley T, editors. Bioprocessing of plant in vitro systems. Cham: Springer International Publishing; 2016. pp. 1–40. [Google Scholar]
  347. Xu L, Chen B, Geng X, et al. A protease-resistant α-galactosidase characterized by relatively acid pH tolerance from the Shitake Mushroom Lentinula edodes. Int J Biol Macromol. 2019;128:324–330. doi: 10.1016/j.ijbiomac.2019.01.051. [DOI] [PubMed] [Google Scholar]
  348. Yang Z, Zhang Z. Engineering strategies for enhanced production of protein and bio-products in Pichia pastoris: a review. Biotechnol Adv. 2018;36:182–195. doi: 10.1016/j.biotechadv.2017.11.002. [DOI] [PubMed] [Google Scholar]
  349. Yang S, Kuang Y, Li H, et al. Enhanced production of recombinant secretory proteins in Pichia pastoris by optimizing Kex2 P1’ site. PLoS ONE. 2013;8:e75347. doi: 10.1371/journal.pone.0075347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Yang H, Wang J, Zhao M, et al. Feasible development of stable HEK293 clones by CRISPR/Cas9-mediated site-specific integration for biopharmaceuticals production. Biotechnol Lett. 2019;41:941–950. doi: 10.1007/s10529-019-02702-5. [DOI] [PubMed] [Google Scholar]
  351. Yao J, Weng Y, Dickey A, Wang K. Plants as factories for human pharmaceuticals: applications and challenges. IJMS. 2015;16:28549–28565. doi: 10.3390/ijms161226122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  352. YaPing W, Ben R, Ling Z, Lixin M. High-level expression of two thermophilic β-mannanases in Yarrowialipolytica. Protein Expr Purif. 2017;133:1–7. doi: 10.1016/j.pep.2017.02.008. [DOI] [PubMed] [Google Scholar]
  353. Yu LX, Amidon G, Khan MA, et al. Understanding pharmaceutical quality by design. AAPS J. 2014;16:771–783. doi: 10.1208/s12248-014-9598-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Yuen KCJ, Johannsson G, Ho KKY, et al (2023) Diagnosis and testing for growth hormone deficiency across the ages: a global view of the accuracy, caveats, and cut-offs for diagnosis. Endocrine Connect. 10.1530/EC-22-0504 [DOI] [PMC free article] [PubMed]
  355. Zeng F, Yang C, Gao X, et al. Comprehensive elucidation of the structural and functional roles of engineered disulfide bonds in antibody Fc fragment. J Biol Chem. 2018;293:19127–19135. doi: 10.1074/jbc.RA118.005367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Zhang J, Lan N. Hirudin variants production by genetic engineered microbial factory. Biotechnol Genet Eng Rev. 2018;34:261–280. doi: 10.1080/02648725.2018.1506898. [DOI] [PubMed] [Google Scholar]
  357. Zhao M, Vandersluis M, Stout J, et al. Affinity chromatography for vaccines manufacturing: finally ready for prime time? Vaccine. 2019;37:5491–5503. doi: 10.1016/j.vaccine.2018.02.090. [DOI] [PubMed] [Google Scholar]
  358. Zhou G, Chen S, Chen Z. Advances in COVID-19: the virus, the pathogenesis, and evidence-based control and therapeutic strategies. Front Med. 2020;14:117–125. doi: 10.1007/s11684-020-0773-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Zhu J. Mammalian cell protein expression for biopharmaceutical production. Biotechnol Adv. 2012;30:1158–1170. doi: 10.1016/j.biotechadv.2011.08.022. [DOI] [PubMed] [Google Scholar]
  360. Zhu MM, Mollet M, Hubert RS, et al. Industrial production of therapeutic proteins: cell lines, cell culture, and purification. In: Kent JA, Bommaraju TV, Barnicki SD, et al., editors. Handbook of industrial chemistry and biotechnology. Cham: Springer International Publishing; 2017. pp. 1639–1669. [Google Scholar]
  361. Zhu W, Xu R, Gong G, et al. Medium optimization for high yield production of human serum albumin in Pichia pastoris and its efficient purification. Protein Expr Purif. 2021;181:105831. doi: 10.1016/j.pep.2021.105831. [DOI] [PubMed] [Google Scholar]
  362. Zhuang H, Hao H, Qiu Y, Gan J, Li T. Recent advances in glycopeptide and glycoprotein synthesis: a refined synthetic probe towards the biological world. Chinese J Chem. 2023;41:2010–2024. doi: 10.1002/cjoc.202300035. [DOI] [Google Scholar]
  363. Zinsli LV, Stierlin N, Loessner MJ, Schmelcher M. Deimmunization of protein therapeutics—recent advances in experimental and computational epitope prediction and deletion. Comput Struct Biotechnol J. 2021;19:315–329. doi: 10.1016/j.csbj.2020.12.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  364. Zydney AL. Continuous downstream processing for high value biological products: a review: continuous downstream processing. Biotechnol Bioeng. 2016;113:465–475. doi: 10.1002/bit.25695. [DOI] [PubMed] [Google Scholar]

Articles from 3 Biotech are provided here courtesy of Springer

RESOURCES