the next generation of venom-based drugs · venom-based drugs sac ˜ briefly explores the potential...

5
28 Prescriber April 2020 prescriber.co.uk RESEARCH B y the pricking of my thumbs, the next generation of venom-based drugs this way comes. You might think that science has come a long way since Macbeth’s witches hubbled and bubbled fillet of a fenny snake, a toad that swelter’d venom and adder’s fork. Yet if you manage heart disease or diabetes, some of your patients will, almost certainly, take ven- om-based drugs. Meanwhile, researchers are developing new venom-based drugs for, among other conditions, autoimmune and cardiovascular diseases, chronic pain and cancer. “Animal-based venoms are hugely promising as an avenue for new drugs,” says Ray Norton, Professor of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Australia. Some venom-based drugs are already therapeutic mainstays. Captopril, the first ACE inhibitor, derives from studies of bradykinin-potentiating peptides isolated from the venom of Bothrops jararaca, a South American pit viper. 1 The venom of the Gila monster (Heloderma suspectum) – found in New Mexico and Arizona and one of the few venomous lizards – con- tains a group of peptides called exendins. These are structurally similar to gluca- gon-like peptide-1 (GLP-1), a human incre- tin hormone that reduces blood glucose. Exenatide, a synthetic version of exendin-4, and other GLP-1 mimetics, are widely used type 2 diabetes treat- ments. 1 Specialists have a wider choice of venom-based drugs (see Table 1). In order to feed, a leech needs to keep the blood flowing. So, leeches evolved a pro- tein called hirudin, which potently inhibits thrombin. A synthetic version of hirudin is available as an anticoagulant. Other ven- om-based drugs alleviate chronic pain and reduce the risk of death and myocar- dial infarction in certain patients with acute coronary syndromes. 1 This article The next generation of venom-based drugs MARK GREENER Some venom-derived drugs, such as captopril and exenatide, are already used therapeutically, and researchers are continuing to develop new venom- based treatments for a wide range of conditions, including autoimmune and cardiovascular diseases, chronic pain and cancer. This article explores the potential for new venom-based drugs and some of the problems researchers need to overcome. Figure 1. Pufferfish skin contains tetrodotoxin, a sodium-channel blocker that is about a thousand times more toxic to humans than cyanide

Upload: others

Post on 21-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The next generation of venom-based drugs · Venom-based drugs SAC ˜ briefly explores the potential for new venom-based drugs and some of the hur - dles facing researchers. An evolving

28 ❚ Prescriber April 2020 prescriber.co.uk

■ RESEARCH

By the pricking of my thumbs, the next generation of venom-based drugs this

way comes. You might think that science has come a long way since Macbeth’s witches hubbled and bubbled fillet of a fenny snake, a toad that swelter’d venom and adder’s fork. Yet if you manage heart disease or diabetes, some of your patients will, almost certainly, take ven-om-based drugs. Meanwhile, researchers are developing new venom-based drugs for, among other conditions, autoimmune and cardiovascular diseases, chronic pain and cancer. “Animal-based venoms are hugely promising as an avenue for new drugs,” says Ray Norton, Professor of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Australia. Some venom-based drugs are already therapeutic mainstays. Captopril, the first ACE inhibitor, derives from studies of bradykinin-potentiating peptides isolated from the venom of Bothrops jararaca, a

South American pit viper.1 The venom of the Gila monster (Heloderma suspectum) – found in New Mexico and Arizona and one of the few venomous lizards – con-tains a group of peptides called exendins. These are structurally similar to gluca-gon-like peptide-1 (GLP-1), a human incre-tin hormone that reduces blood glucose. Exenatide, a synthetic version of exendin-4, and other GLP-1 mimetics, are widely used type 2 diabetes treat-ments.1

Specialists have a wider choice of venom-based drugs (see Table 1). In order to feed, a leech needs to keep the blood flowing. So, leeches evolved a pro-tein called hirudin, which potently inhibits thrombin. A synthetic version of hirudin is available as an anticoagulant. Other ven-om-based drugs alleviate chronic pain and reduce the risk of death and myocar-dial infarction in certain patients with acute coronary syndromes.1 This article

The next generation of venom-based drugsMARK GREENER

Some venom-derived drugs, such as captopril and exenatide, are already used therapeutically, and researchers are continuing to develop new venom-based treatments for a wide range of conditions, including autoimmune and cardiovascular diseases, chronic pain and cancer. This article explores the potential for new venom-based drugs and some of the problems researchers need to overcome.

Figure 1. Pufferfish skin contains tetrodotoxin, a sodium-channel blocker that is about a thousand times more toxic to humans than cyanide

Page 2: The next generation of venom-based drugs · Venom-based drugs SAC ˜ briefly explores the potential for new venom-based drugs and some of the hur - dles facing researchers. An evolving

Prescriber April 2020 ❚ 29prescriber.co.uk

Venom-based drugs l RESEARCH ■

briefly explores the potential for new venom-based drugs and some of the hur-dles facing researchers.

An evolving ideaVenom seems to have evolved about 90 times across the animal kingdom and almost 15% of animal species seem to be venomous.2 Ron Jenner, Research Leader in the Division of Invertebrates at London’s Natural History Museum, notes that many venoms disrupt the nervous system or blood-vascular sys-tem to quickly disable a prey or cause pain that deters further attack. So, a venom can give an animal a marked sur-vival advantage. “A non-venomous rep-tile may be able to catch and overcome prey with claws and teeth alone, but if it is able to inject a venom at the same time to help disable prey more quickly, it may be selected for in evolution,” he says. “The adaptive value is illustrated by venoms having evolved dozens of times in the animal kingdom, and by the fact that once venom has evolved it is rarely or never lost.” Indeed, venoms emerged early in evo-lution of life on earth. The Burgess Shale, which is about 505 million years old, probably contains fossilised remains of early venomous animals. “The earliest venomous animals are probably ancient relatives of jellyfish and sea anemones, a group known as Cnidaria,” remarks Dr Jenner, co-author of Venom: The Secrets of Nature’s Deadliest Weapon,2 an acces-sible introduction to this fascinating area. “The Burgess Shale contains fos-sils of ancient cnidarian relatives. Although we don’t know if they were ven-omous, they could very well have been.” Over the millennia, some animals developed exquisitely toxic venoms. A single bite of a coastal taipan (Oxyuranus scutellatus) could kill 3.4 million mice – or 1000 adult humans.2 Being venomous means more than just being toxic. The animal must make the toxin in special-ised cells and have a specific delivery mechanism.2,3 The skin of the pufferfish (Tetraodontidae sp.) contains tetrodo-toxin, which is some thousand times more toxic to humans than cyanide (see Figure 1).4 So, predators spit the puffer-fish out.

Venomous animals actively deliver toxins. The Gila monster produces venom in glands in its lower jaw, which travels along grooved teeth and into the victim as the lizard chews.1 “Mouthparts like teeth have evolved into sophisticated venom delivery structures in reptiles and some groups of mammals, which have grooved or tube-like teeth to deliver venom more effectively,” Dr Jenner com-ments. “Pointy spines in fish, which were probably already used as defence struc-tures, have, in some cases, evolved into sophisticated syringe-like venom injec-tion machinery.”

Toxic themesThe earliest centipedes seem to have hunted using venoms containing four tox-ins. Over the last 400 million years, cen-tipedes seem to have evolved 93 distinct families of peptides and proteins as tox-ins that show “astonishing structural

diversity”.5 “This structural and func-tional diversity is true for the venoms of other taxa, such as snakes and various groups of venomous insects,” Dr Jenner says. “Nevertheless, some themes have emerged. Many venoms target the physi-ological Achilles heel of victims: the nerv-ous and vascular systems. Toxins in venoms targeting the former often modu-late ion channels or neurotransmitter receptors; those affecting the latter often target the blood clotting cascade. The ecologically relevant time for the action of venoms is usually seconds to a few minutes – if it takes longer, prey will have escaped or a predator may already have eaten its prey. So, venoms evolved to tar-get the nervous and vascular systems to have a rapid effect.” As venoms disrupt essential physio-logical functions, toxins have proved to be invaluable pharmacological tools. In the early 1960s, researchers discovered

Generic name Species Examples of indication

Bivalirudin Medicinal leech (Hirudo medicinalis)

Anticoagulant for adults undergoing percutaneous coronary intervention (PCI); adults with unstable angina/ non-ST segment elevation myocardial infarction (MI) planned for urgent or early intervention

Captopril Jararaca pit viper (Bothrops jararaca)

Hypertension, congestive heart failure, MI, type I diabetic nephropathy

Eptifibatide Pigmy rattlesnake (Sistrurus miliarius barbouri)

Prevention of early MI in adults presenting with unstable angina or non-Q-wave MI

Exenatide Gila monster (Heloderma suspectum)

Adults with type 2 diabetes mellitus

Tirofiban Saw-scaled viper (Echis carinatus)

Prevention of early MI in adults presenting with acute coronary syndromes without ST elevation; reduction of major cardiovascular events in patients with acute ST elevation MI (STEMI) intended for primary PCI

Ziconotide Cone snail (Conus magus)

Severe, chronic pain in adults who require intrathecal analgesia

Based on summaries of product characteristics and Jenner and Undheim2

Table 1. Currently available venom-based drugs, the species they were originally sourced from and their indications

Page 3: The next generation of venom-based drugs · Venom-based drugs SAC ˜ briefly explores the potential for new venom-based drugs and some of the hur - dles facing researchers. An evolving

■ RESEARCH l Venom-based drugs

30 ❚ Prescriber April 2020 prescriber.co.uk

that alpha-bungarotoxin, isolated from the venom of the Taiwanese banded krait snake (Bungarus multicinctus), blocks postsynaptic neuromuscular transmis-sion in higher ver tebrates. Alpha-bungarotoxin allowed researchers to discriminate between and physically sep-arate nicotinic receptors and acetyl-cholinesterase, both of which bind acetylcholine.6 “We still need pure venom-derived molecules as molecular probes,” says Professor Nor ton. “Displacement studies of known ligands for a particular target can be an efficient route to novel venom-derived ligands (peptides or otherwise) for that target.” For example, a species of European bloodworm (Glycera tridactyla) causes massive, chronic, but reversible, acetyl-choline release into the neuromuscular junction. This allows researchers to explore cholinergic transmission.2 Indeed, some toxins are remarkably specific. Over 400 peptide toxins target sodium channels, including more than 182 from scorpions, 116 from spiders and 55 from sea anemones.2 Tetro-dotoxin also blocks sodium channels,4 which is why fugu (pufferfish) can, if poorly prepared, kill adventurous gastro-nauts. “Many neurotoxic peptides show great affinity for particular types of ion channels,” Dr Jenner says. “By binding to these specific ion channels, neurotoxins can stimulate or block nerve impulses, which can lead to whole body paralysis.” Ion channels are ubiquitous in the ani-mal kingdom. But that’s no guarantee that a venom will work in humans. “Neurotoxins present in venoms from snakes in the cobra family (Elapidae) have specific molecular targets in nerves and at the neuromuscular junction. So they can par-alyse a wide range of animals, including humans,” Dr Jenner says. “But ion chan-nels can vary considerably between ani-mal groups. A neurotoxin in the venom of a bird-eating snake may have much more powerful effects on bird ion channels than mammal ion channels.” Other venom components have non-specific effects. The sea wasp jelly-fish (Chironex fleckeri) delivers a venom that triggers inflammation and is toxic to nerves, blood, cells, the heart, muscle and skin.2 “Many animal venoms, includ-

ing fish and snake venoms, contain the enzyme hyaluronidase,” Dr Jenner says. “Hyaluronidase breaks down the extracel-lular matrix, and can thereby act as a spreading factor that allows venom tox-ins to better penetrate the bodies of envenomated victims.” Incidentally, hyalu-ronidase may increase the penetration of chemotherapy to some malignancies, including certain pancreatic cancers.7 “Other venom components, like the so-called pore-forming toxins, are also relatively non-specific,” Dr Jenner says. Indeed, pore-forming peptides from the venom of some species of scorpion, such as the African yellow leg scorpion (Opistophthalmus carinatus) or the bur-rowing thick tail scorpion (Parabuthus schlechteri), seem effective against sev-eral Gram-negative bacteria including Escherichia coli, Pseudomonas aerugi-nosa and Haemophilus influenzae (see Figure 2).8

New venom-based drugsThe toxin’s effect can offer a clue to pos-sible clinical applications. If you’re male, in Brazil and unfortunate enough to be

b i t ten by the wander ing spider (Phoneutria nigriventer) or yellow scor-pion (Tityus serrulatus), you’ll probably suffer tachycardia, sweating, excessive salivation and vomiting. If that wasn’t enough, you could experience spontane-ous erections. So, these species’ ven-oms are being investigated as a possible treatment for erectile dysfunction that doesn’t respond to sildenafil or other existing drugs.2,9 The cone snail (Conus geographus) – the world’s deadliest snail – induces hypoglycaemia to sedate fish before cap-ture. C. geographus also kills humans by causing paralysis with, Professor Norton says, “a cocktail of neuro- and myo- toxins”.2 “Our work on cone snail-derived insulins also offers hope for new insulin analogues to treat diabetes,” Professor Norton adds. Indeed, the seas seem to be rich sources of pharmaceutically active tox-ins. The Caribbean sun anemone (Stichodactyla helianthus), for instance, produces a toxin that strongly blocks several potassium channels on T lym-phocytes, the brain and cardiac tissues.

Figure 2. Pore-forming peptides from the venom of some species of scorpion (such as Opistophthalmus carinatus, shown) may be effective against several Gram-negative bacteria including Escherichia coli, Pseudomonas aeruginosa and Haemophilus influenzae

SPL

Page 4: The next generation of venom-based drugs · Venom-based drugs SAC ˜ briefly explores the potential for new venom-based drugs and some of the hur - dles facing researchers. An evolving

Prescriber April 2020 ❚ 31prescriber.co.uk

Venom-based drugs l RESEARCH ■

Modifying this toxin produced dalaza-tide, which is 100 times more selective for potassium channels on T lympho-cytes than those on brain and cardiac cells. Dalazatide is showing promise in several autoimmune diseases including multiple sclerosis, rheumatoid arthritis and psoriasis.1,10,11 Professor Norton’s laboratory is developing HsTX1[R14A], isolated from the giant forest scorpion (Heterometrus spinifer), which blocks voltage-gated potassium channels, for autoimmune dis-eases and other conditions. In a rat mode l o f rheumato id a r th r i t i s , HsTX1[R14A] was effective when admin-istered weekly. PEGylation may further increase the duration of action, perhaps allowing fortnightly administration.12,13

Venom-based drugs could also lead to a new generation of cancer treat-ments. A toxin from one of the few ven-omous mammals, the nor thern Short-tailed shrew (Blarina brevicauda), inhibits growth of ovarian and breast can-cer in animal models as well as showing promising activity and tolerability in pre-liminary studies of advanced epithelial tumours.1 Chlorotoxin, isolated from the venom of the deathstalker scorpion (Leiurus quinquestriatus hebraeus), blocks chlo-ride channels. Chlorotoxin also inhibits and lowers expression of a certain sub-type (MMP-2) of the enzyme matrix metal-loprotease. Gliomas and some other cancer cells express high levels of MMP-2, which is not normally expressed by brain cells.1,8 MMP-2 degrades and remodels extracellular matrix, which con-tributes to the growth and spread of the cancer.8 Chlorotoxin could target radio-chemicals and imaging agents to the tumour to treat or diagnose certain can-cers. One approach attaches a fluores-cent dye to chlorotoxin. So, the surgeon can see the cancer in the operating room and remove as much of the tumour as possible.1

Antivenoms and pesticidesSnakes envenomate 1.8 million people a year worldwide. About 100,000 people die. And, as many snake venoms cause massive tissue destruction, some 400,000 people need amputations.

Scorpions and spiders kill another 5000 people.2 Antivenoms are antibodies that neutralise the toxin. However, antiven-oms are highly species-specific and can induce severe immunological reactions and other side-effects in between 43% and 81% of patients.3 The cost also puts anti-venoms out of reach of many people in the poorest parts of the world, who are the most likely to be envenomated.2

The growing understanding of the pharmacological actions of venoms offers new treatments for this major pub-lic health problem. The enzyme sphingo-myelinase D is largely responsible for severe tissue necrosis and systemic effects than can follow a bite from a recluse spider (Loxosceles sp. ) . Researchers developed small molecules that inhibit sphingomyelinase D. Some of these inhibited the enzyme’s binding to human keratinocytes and reduced venom-induced cell death and the devel-opment of necrotic lesions in rabbits.14 “This is an interesting area of develop-ment,” Professor Norton says. “For exam-ple, if the harmful effects of a particular venom can be attributed to a particular enzyme – such as phospholipase, which is common in snake venoms – an inhibi-tor could be a useful treatment for enven-omation. It’s important that the inhibitor be specific for the venom, as opposed to the endogenous, enzyme.” “The more we understand about the composition of a venom and the actions of its toxins individually and in combina-tion, the better chances we have of developing treatments for envenoma-tion,” Dr Jenner adds. “Many animals are resistant in varying degrees to snake venoms, including venomous snakes themselves, their prey, and their predators. For instance, many vipers have haemorrhagic metalloprotease enzymes in their venom. Some preda-tors of rattlesnakes, such as opossums, have serum proteins that neutralise the enzymes.” Many animals produce toxins to sub-due insect prey. Thus, venom-based insecticides could help tackle the 1000 or so insect pests that reduce the world’s crop production by an estimated 10–14%. One peptide-based insecticide derives from the venom of the Australian Blue

Moun ta i ns f unne l web sp i de r (Hadronyche versuta), which is about 10,000-fold more selective for voltage- dependent calcium channels in insects than other vertebrates.1 “I believe this is a very promising avenue of development for venom-derived peptides,” comments Professor Norton.

Untangling toxinsDespite the success of captopril and the promise of sphingomyelinase D inhibi-tors, developing orally active venom- based drugs (peptidomimetics) is diffi-cult. So, venom-based drugs will, initially at least, probably reach the market as peptides. “Peptides are becoming cheaper and easier to make, and there are several options for delivering pep-tides via non-oral routes, including inhaled, slow-release and transdermal formulations,” Professor Norton notes. “Small drug-like compounds generally cannot match the potency and target specificity of peptides, which typically have a much larger contact surface area with their receptors. We and others have tried designing peptidomimetics, but with limited success. Nonetheless, molecular design continues to advance, and, com-bined with screening of chemical librar-ies, offers the prospect of small-molecule mimetics of venom peptides in the future.” To complicate matters further, venoms are, typically, complex chemical cocktails. The venom of a cone snail or spider can contain hundreds or thousands of differ-ent proteins.2 And the components often act in concert. For instance, the venom of the Chinese red-headed centipede (Scolopendra subspinipes) is 3500 to 50,000 times more insecticidal than the neurotoxins tested alone.2

“There are examples where activity- driven fractionation of a venom falls off a cliff because the activity declines or dis-appears as purification proceeds,” Professor Norton says. “Venom toxins work together to disrupt normal physio-logical functioning,” Dr Jenner adds. “In some cases, we understand exactly how venom components interact. But because venoms can comprise hundreds of distinct chemicals, there is still much to be learned about the toxic synergy of

Page 5: The next generation of venom-based drugs · Venom-based drugs SAC ˜ briefly explores the potential for new venom-based drugs and some of the hur - dles facing researchers. An evolving

■ RESEARCH l Venom-based drugs

32 ❚ Prescriber April 2020 prescriber.co.uk

venom components.” Researchers face other challenges, including getting enough venom to study. “Isolating active peptides is straightfor-ward with modern chromatographic meth-ods,” Professor Norton comments. “A bigger challenge is getting sufficient venom to begin fractionation.” Venom may account for 0.5% of a snake’s or scorpion’s body weight. But refilling the venom glands can mean raising the met-abolic rate by 20% in a snake and up to 40% in some scorpions.2 As manufactur-ing venoms is metabolically expensive, most animal produce small amounts. An approach called ‘venomics’ – which inte-grates proteomics, genomics and tran-scriptomics (study of RNA derived from genes) – allows comprehensive analyses of venoms and should help identify potential leads for new drugs, despite the limited quantities of venom.15

Indeed, researchers have barely scratched the surface of venoms’ phar-maceutical potential. There are, for example, more than 3100 described spe-cies of centipedes across five orders. But most of the research, including that for new drugs, focuses on a few species from a single family (Scolopendridae).5 More than 2900 fish species use venom as a defence,16 but Dr Jenner points out most are poorly studied. “There are probably more than 200,000 species of living venomous ani-mals, the vast majority of which have not yet had their venoms profiled,” Dr Jenner says. “Each is chock-full with bioactive compounds, and a number of them have already been used to develop pharma-ceuticals. However, the route from a

promising venom toxin to a drug is long, arduous, expensive, and frequently a dead end.” “Genomic, transcriptomic and pro-teomic studies of venom are identifying thousands of, mainly, new peptides, some of which can be turned into valua-ble therapeutic leads,” Professor Norton concludes. “Our challenge is to screen these candidates efficiently in order to kiss as few frogs as possible before find-ing the princes.”

References1. Pennington MW, et al. Peptide therapeutics from venom: current status and potential Bioorg Med Chem 2018;26:2738–58.2. Jenner R, Undheim E. Venom: the secrets of nature’s deadliest weapon. London: National History Museum, 2017.3. Wilcox C. Venomous: how earth’s deadliest creatures mastured biochemisty. New York: Scientific American, 2017.4. Bane V, et al. Tetrodotoxin: chemistry, toxic-ity, source, distribution and detection. Toxins (Basel) 2014;6:693–755.5. Jenner RA, et al. Parallel evolution of com-plex centipede venoms revealed by compara-tive proteotranscriptomic analyses. Mol Biol Evol 2019; DOI: 10.1093/molbev/msz1816. Changeux J-P The nicotinic acetylcholine receptor: the founding father of the pen-tameric ligand-gated ion channel superfamily. J Biol Chem 2012;287:40207–15.7. van Mackelenbergh MG, et al. Clinical trials targeting the stroma in pancreatic cancer: a systematic review and meta-analysis. Cancers 2019;11:588.8. Shah PT, et al. Scorpion venom: a poison or a medicine-mini review. Indian J Geo-Marine Sci 2018;47:773–8.9. Nunes KP, et al. Erectile function is improved in aged rats by PnTx2-6, a toxin from

Phoneutria nigriventer spider venom. J Sexual Med 2012;9:2574–81.10. Prentis P, et al. Sea anemones: quiet achievers in the field of peptide toxins. Toxins 2018;10:36.11. Tarcha EJ, et al. Safety and pharmacody-namics of dalazatide, a Kv1.3 channel inhibi-tor, in the treatment of plaque psoriasis: a randomized phase 1b trial. PLOS ONE 2017;12:e0180762.12. Bergmann R, et al. Distribution and kinetics of the Kv1.3-blocking peptide HsTX1[R14A] in experimental rats. Sci Rep 2017;7:3756.13. Tanner MR, et al. Prolonged immunomod-ulation in inflammatory arthritis using the selective Kv1.3 channel blocker HsTX1[R14A] and its PEGylated analog. Clin Immunol 2017;180:45–57.14. Lopes PH, et al. Targeting Loxosceles spi-der sphingomyelinase D with small-molecule inhibitors as a potential therapeutic approach for loxoscelism. J Enzyme Inhib Med Chem 2019;34:310–21.15. Wilson D, Daly NL. Venomics: a mini-re-view. High-Throughput 2018;7. DOI: 10.3390/ht703001916. Harris RJ, Jenner RA. Evolutionary ecology of fish venom: adaptations and consequences of evolving a venom system. Toxins 2019;11:60.

Declaration of interestsMark Greener is a full-time medical writer and journalist and, as such, regularly pro-vides editorial and consultancy services to numerous pharmaceutical, biotechnol-ogy and device companies and their agencies. He has no shares or financial interests relevant to this article.

Mark Greener is a freelance medical writer and journalist