Biopharmaceuticals

Chapter 12 Biopharmaceuticals




Introduction


The term ‘biopharmaceutical’ was originally coined to define therapeutic proteins produced by genetic engineering, rather than by extraction from normal biological sources. Its meaning has broadened with time, and the term now encompasses nucleic acids as well as proteins, vaccines as well as therapeutic agents, and even cell-based therapies. In this chapter we describe the nature of biopharmaceuticals, and the similarities and differences in discovery and development between biopharmaceuticals and conventional small-molecule therapeutic agents. The usual starting point for biopharmaceuticals is a naturally occurring peptide, protein or nucleic acid. The ‘target’ is thus identified at the outset, and the process of target identification and validation, which is a major and often difficult step in the discovery of conventional therapeutics (see Chapter 6), is much less of an issue for biopharmaceuticals. Equally, the process of lead finding and optimization (Chapters 7, 8, 9) is generally unnecessary, or at least streamlined, because nature has already done the job. Even if it is desirable to alter the properties of the naturally occurring biomolecule, the chemical options will be much more limited than they are for purely synthetic compounds. In general, then, biopharmaceuticals require less investment in discovery technologies than do conventional drugs. Toxicity associated with reactive metabolites – a common cause of development failure with synthetic compounds – is uncommon with biopharmaceuticals. On the other hand, they generally require greater investment in two main areas, namely production methods and formulation. Production methods rely on harnessing biological systems to do the work of synthesis, and the problems of yield, consistency and quality control are more complex than they are for organic synthesis. Formulation problems arise commonly because biomolecules tend to be large and unstable, and considerable ingenuity is often needed to improve their pharmacokinetic properties, and to target their distribution in the body to where their actions are required.


It is beyond the scope of this book to give more than a brief account of the very diverse and rapidly developing field of biopharmaceuticals. More detail can be found in textbooks (Buckel, 2001; Ho and Gibaldi, 2003; Walsh, 2003). As the field of biopharmaceuticals moves on from being mainly concerned with making key hormones, antibodies and other signalling molecules available as therapeutic agents, efforts – many of them highly ingenious – are being made to produce therapeutic effects in other ways. These include, for example, using antisense nucleic acids, ribozymes or RNAi (see below and Chapter 6) to reduce gene expression, the use of catalytic antibodies to control chemical reactions in specific cells or tissues, and the development of ‘DNA vaccines’. So far, very few of these more complex ‘second-generation’ biopharmaceutical ideas have moved beyond the experimental stage, but there is little doubt that the therapeutic strategies of the future will be based on more sophisticated ways of affecting biological control mechanisms than the simple ‘ligand → target → effect’ pharmacological principle on which most conventional drugs are based.



Recombinant DNA technology: the engine driving biotechnology


The discovery of enzymes for manipulating and engineering DNA – the bacterial restriction endonucleases, polynucleotide ligase and DNA polymerase – and the invention of the enabling technologies of DNA sequencing and copying of DNA sequences by using the polymerase chain reaction (PCR) allowed rapid determination of the amino acid sequence of a protein from its mRNA message. Versatile systems for introducing nucleic acids into target cells or tissues and for the control of host nucleic acid metabolism brought the potential for correction of genetic defects and for new therapeutic products for disorders poorly served by conventional small-molecule drugs. The importance of these discoveries for the biological sciences was indicated by the many Nobel Prizes awarded for related work. No less critical was the impact that these reagents and technologies had on applied science, especially in the pharmaceutical industry. The business opportunities afforded by biotechnology spawned thousands of startup companies and profoundly changed the relationship between academia and industry. The mainstream pharmaceutical industry, with its 20th-century focus on small-molecule therapeutic agents, did not immediately embrace the new methodologies except as research tools in the hands of some discovery scientists. Entrepreneurs in biotechnology startup firms eventually brought technology platforms and products to large pharmaceutical companies as services or as products for full-scale development. This alliance allowed each party to concentrate on the part they did best.


Biotechnology products were naturally attractive to the small startup companies. Because the protein or nucleic acid itself was the product, it was unnecessary to have medicinal chemists synthesize large collections of organic small-molecule compounds to screen for activity. A small energetic company with the right molecule could come up with a profitable and very useful product. The niche markets available for many of the initial protein or nucleic acid products were sufficient to support a small research-based company with a high-profit-margin therapeutic agent.



The early days of protein therapeutics


Along with plant-derived natural products, proteins and peptides were some of the first therapeutic agents produced by the fledgling pharmaceutical industry in the latter half of the 19th century, before synthetic chemistry became established as a means of making drugs. Long before antibiotics were discovered, serum from immune animals or humans was successfully used to treat a variety of infectious diseases. Serotherapy was the accepted treatment for Haemophilus influenzae meningitis, measles, diphtheria, tetanus, hepatitis A and B, poliovirus, cytomegalovirus and lobar pneumonia. Antisera raised in animals were used to provide passive protection from diphtheria and tetanus infection.


Extracts of tissues provided hormones, many of which were polypeptides. After its discovery in 1921, insulin extracted from animal pancreas replaced a starvation regimen for treating diabetes. The size of the diabetic population, and the activity in humans of the hormone isolated from pancreas of pigs and cows, permitted early commercial success. Generally, however, the low yield of many hormones and growth factors from human or animal sources made industrial scale isolation difficult and often uneconomic. Nevertheless, several such hormones were developed commercially, including follicle-stimulating hormone (FSH) extracted from human urine to treat infertility, glucagon extracted from pig pancreas to treat hypoglycaemia, and growth hormone, extracted from human pituitary to treat growth disorders. Some enzymes, such as glucocerebrosidase, extracted from human placenta and used to treat an inherited lipid storage disease (Gaucher’s disease), and urokinase, a thrombolytic agent extracted from human urine, were also developed as commercial products.


Some serious problems emerged when proteins extracted from human or animal tissues were developed for therapeutic use. In particular:



The recombinant DNA revolution and the subsequent development of biotechnology resolved many of these issues. Many vaccines and antisera, however, are still prepared from blood products or infectious organisms, rather than by recombinant DNA methods.



Currently available classes of biopharmaceuticals


The major classes of biopharmaceuticals currently on the market include hormones, cytokines, growth factors, antibodies, enzymes, vaccines and nucleotide-based agents. Examples of therapeutic proteins, including antibodies, enzymes and other proteins approved for clinical use, are presented in Table 12.1. Others not included in the compilation include therapeutic preparations such as serum albumin, haemoglobin and collagen, which are not drugs in the conventional sense.



In addition to the ‘mainstream’ biopharmaceuticals considered here are numerous speciality products for niche markets that are under investigation or in development by small ‘boutique’ companies.



Growth factors and cytokines


The production, differentiation and survival of the various types of blood cell are tightly regulated by an interacting network of hormones, cytokines and growth factors. Species-specific activities of many of these chemical mediators, and their very low abundance, highlighted the need for biopharmaceutical products.


The most common uses of haemopoietic factors are for the treatment of various types of neutropenia, where specific white cell levels are depressed as a result of infection, immune disorders, recovery from chemotherapy or reaction to various drug regimens. They are especially useful in aiding recovery from the dose-limiting side effects of cancer chemotherapy. Granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) are used for this purpose to improve patient quality of life and allow the continuation of chemotherapy.


Erythropoietin (EPO), normally secreted by the kidney to stimulate the production of red blood cells, is the most successful biotechnology product so far marketed. EPO boosts red cell counts and reduces transfusion requirements for patients rendered anaemic by cancer chemotherapy or renal disease. Various forms of EPO with clearance profiles – and hence duration of action – modified by linkage with polyethylene glycol (PEGylation) or alteration of its glycosylation (see below and Chapter 17) are also available. Off-label use of EPO by athletes to improve performance has caused controversy.


Interferons are a complex group of proteins that augment immune effector cell function. Interferon-α was the first recombinant biotherapeutic agent approved by the FDA for cancer treatment. Recombinant interferons have been approved for melanoma, hepatitis C, Karposi’s sarcoma, T-cell lymphoma, chronic myelogenous leukaemia, multiple sclerosis and severe malignant osteopetrosis. Mechanism-based side effects have thus far restricted their utility to these severe disorders.



Hormones


Hormone replacement or augmentation is a commonly accepted medical practice in certain diseases of deficiency or misregulation. Insulin isolated from biological sources is administered to diabetics to control blood glucose levels. Immunological reaction to non-human (porcine or bovine) insulin preparations, which occur in a significant number of patients, are avoided by recombinant products incorporating parts of the human insulin sequence. A variety of human insulin and glucagon preparations are now on the market.


Human growth hormone (somatotropin) was originally developed for treating paediatric growth failure and Turner’s syndrome. Originally, growth hormone was extracted from human pituitary tissue post mortem, but this material carried a significant risk of transmitting Creutzfeld-Jakob disease, a fatal neurodegenerative condition now known to be transmitted by a prion, an abnormal protein found in affected nervous tissue, whose existence was unsuspected when human-derived growth hormone was introduced as a therapeutic agent. The production of human growth hormone by recombinant methods rather than extraction avoids this serious problem as well as providing a much more abundant source. Growth hormone has acquired notoriety since the potential for misuse to produce taller and stronger athletes was realized.


Human gonadotropin-releasing hormones have been extensively used in fertility management as well as in treating endometriosis and precocious puberty. Originally isolated from urine, there are now numerous recombinant products on the market.





Therapeutic antibodies



Monoclonal antibodies


A breakthrough in high-quality reproducible and scalable production of antibodies came with the development by Kohler and Milstein in 1975 of monoclonal antibodies. Fusion of primed T cells from an immunized mouse with an immortalized mouse myeloma (B-cell) line that secretes immunoglobulin light chains provided a cell-culture system that could produce unlimited quantities of antibody with defined specificity. Single-cell clones secreted antibody against a single epitope of the antigen. The initial immunization could, for toxic or scarce immunogens, be replaced by in vitro stimulation of isolated mouse thymocytes for fusion with the myeloma cells.


The technology for antibody production has now gone mouseless. It has moved into the realm of molecular biology, in which bacteriophages, gene libraries in plasmids and bacterial hosts are engineered to produce either whole antibodies or derivatives of antibodies with desired properties. ‘Humanizing’ the antibodies, or replacing the rodent constant domains with human sequences (chimerization), limits hypersensitivity reactions to foreign protein. Chimerization increases the half-life of the antibodies in human plasma up to six-fold and improves their function within the human immune network. The human Fc domain reacts with Fc receptors on human cells more avidly. Chimeric antibodies with human constant regions also interact optimally with human complement proteins, and are thus more effective in destroying target cells in patients than are their rodent counterparts.



Antibody selection by phage display


Phage display technology (Benhar, 2001) is a useful way to identify antigen combining regions to produce monoclonal antibodies that bind to therapeutically relevant antigens. Bacteriophages (or phages) are viruses that replicate in bacteria, Escherichia coli being the organism of choice in most cases. For antibody selection (Figure 12.1) a large DNA library, encoding millions of different putative antigen-binding domains, is incorporated into phage DNA, so that each phage particle encodes a single antigen combining region. The mixed phage population is added to E. coli cultures, where the phage replicate, each phage particle expressing copies of a single antigen combining region on its surface. The phage suspension is applied to plates coated with the antigen of interest (‘panning’) and those phage particles expressing antigen combining regions recognizing the antigen stick to the plates. The adherent phages are isolated, allowing the antigen combining region-encoding DNA to be identified and inserted into the DNA sequence encoding an appropriate full-size antibody scaffold to produce a specific antibody, or into a reduced size, simplified monomeric framework to produce a single-chain (sFv) antibody.




Uses of antibodies as therapeutic agents



Cancer immunotherapy


Although high-affinity mouse monoclonal antibodies to target antigens can be reproducibly produced in industrial quantities, and bind to specific human targets, they generally function poorly in recruiting human effector functions. The therapeutic potency of these antibodies can be enhanced by taking advantage of the targeting selectivity of antibodies (see Chapter 17) for the purposes of drug delivery. Linking other agents, such as cytotoxic drugs, biological toxins, radioisotopes or enzymes to activate prodrugs to targeting antibodies enhances their delivery to the target cells and reduces side effects by directing the toxic agents to the tumour and minimizing clearance. Bispecific antibodies with one H-L chain pair directed against a target cell antigen and the other against a soluble effector such as a complement component, or against a cell surface marker of an effector cell type, have also been developed to bring the components of the reaction together. A number of these are in clinical trials for a variety of different malignancies, but have in general proved less successful than had been expected on the basis of animal studies.




Catalytic antibodies


Antibodies can be used to enhance the chemical reactivity of molecules to which they bind. Such catalytic antibodies (‘abzymes’) created with transition state analogs as immunogens specifically enhance substrate hydrolysis by factors of 102–105 over the rate in their absence, and this principle has been applied to the development of therapeutic agents (Tellier, 2002). Both esterase and amidase activities have been reported. Catalytic turnover of substrates by abzymes is low in comparison to true enzymes, as high-affinity binding impedes the release of products. Attempts to improve catalytic efficiency and to identify therapeutic uses for catalytic antibodies have engrossed both academic and biotech startup laboratories. Targets being approached with these antibodies include cocaine overdose and drug addiction, bacterial endotoxin, and anticancer monoclonal antibody conjugated with a catalytic antibody designed to activate a cytotoxic prodrug. Attempts are also being made to develop proteolytic antibodies containing a catalytic triad analogous to that of serine proteases, designed to cleave gp120 (for treatment of HIV), IgE (for treatment of allergy), or epidermal growth factor receptor (for treatment of cancer).




Vaccines


Using the immune system to protect the body against certain organisms or conditions is a powerful way to provide long-term protection against disease. Unlike with small-molecule pharmaceuticals, which are administered when needed, once immunity is present subsequent exposure to the stimulus automatically activates the response. Most current vaccines are against disease-causing organisms such as bacteria, viruses and parasites. More complex conditions where the antigens are not so well defined are also being addressed by immunization. Vaccines are being tested for cancer, neurodegenerative diseases, contraception, heart disease, autoimmune diseases, and alcohol and drug addiction (Rousseau et al., 2001; Biaggi et al., 2002; BSI Vaccine Immunology Group, 2002; Kantak, 2003). Immune induction is complex. Pioneering experiments with attenuation of disease organisms showed that illness was not required for immunity. The goal in immunization is to retain enough of the disease-causing trait of an antigen to confer protection without causing the disease. For infectious agents, various methods of killing or weakening the organism by drying or exposure to inactivating agents still dominate manufacturing processes. Isolation of antigens from the organisms, or modifying their toxins, can be used in some cases. Vaccine production of isolated antigen ‘subunit’ vaccines benefits from protein engineering.


Novel approaches to presenting antigens are expected to have an impact on vaccination. An example is the phage display technique (Benhar, 2001), described earlier as a technique for antibody selection. The same approach can be used to provide the protein antigen in a display framework that enhances its immunogenicity and reduces the requirement for immune adjuvants (of which there are few approved for human use). Viruses encoding multiple antigens (‘vaccinomes’) can also be employed.


Genetic vaccination (Liu, 2003) employs a DNA plasmid containing the antigen-encoding gene, which is delivered to the host tissue by direct injection of DNA, or by more exotic techniques such as attaching the DNA to microparticles which are shot into tissues at high speed by a ‘gene gun’, or introduced by other transfection methods (Capecchi et al., 2004; Locher et al., 2004; Manoj et al., 2004). When the DNA is transcribed, the mRNA translated and the protein expressed in the host tissue, eukaryotic sequences will undergo appropriate post-translational modification, which does not occur with conventional protein vaccines. In general, partial sequences of pathogen-derived proteins are fully immunogenic, despite lacking the toxicity of full-length transcripts. The first human trial for a genetic vaccine against HIV took place in 1995, and others quickly followed, including hepatitis, influenza, melanoma, malaria, cytomegalovirus, non-Hodgkin’s lymphoma, and breast, prostate and colorectal tumours. Initial results have been promising. At the time of writing, recombinant vaccines against hepatitis A and B (e.g. Twinrix), papillomavirus virus for genital warts and cervical cancer (Gardisil), and against Haemophilus b/meningococcal protein for meningitis (Comvax) are approved. Single or multiple proteins from an organism can be included, and proteins that interfere with the immune response (common in many infectious agents) excluded.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 1, 2016 | Posted by in GENERAL SURGERY | Comments Off on Biopharmaceuticals

Full access? Get Clinical Tree

Get Clinical Tree app for offline access