throbber
Chem Soc Rev
`
`TUTORIAL REVIEW
`
`Cite this: Chem. Soc. Rev., 2021,
`
`50, 1480
`
`Peptides as a platform for targeted therapeutics
`for cancer: peptide–drug conjugates (PDCs)
`
`a Daniel H. O’ Donovan,
`a Jessica Iegre,
`Bethany M. Cooper,
`Maria O¨ lwegård Halvarssonc and David R. Spring
`*a
`
`b
`
`Peptides can offer the versatility needed for a successful oncology drug discovery approach. Peptide–
`drug conjugates (PDCs) are an emerging targeted therapeutic that present increased tumour penetration
`and selectivity. Despite these advantages, there are still limitations for the use of peptides as therapeutics
`exemplified through their slow progression to get into the clinic and limited oral bioavailability. New
`approaches to address these problems have been studied and successfully implemented to enhance the
`stability of peptides and their constructs. There is great promise for the future of PDCs with two
`molecules already on the market and many variations currently undergoing clinical trials, such as
`bicycle-toxin conjugates and peptide–dendrimer conjugates. This review summarises the entire process
`needed for the design and successful development of an oncology PDC including chemical and nano-
`material strategies to enhance peptide stability within circulation, the function of each component of a
`PDC construct, and current examples in clinical trials.
`
`Received 30th September 2020
`
`DOI: 10.1039/d0cs00556h
`
`rsc.li/chem-soc-rev
`
`Key learning points
`1. A variety of methods to address peptide chemical and enzymatic stability can be implemented.
`2. The design of a PDC requires understanding of the mechanism of action intended and hence environmental stimuli (pH, GSH and enzymes).
`3. The function and rationale behind the design of each component of a PDC.
`4. The stability of the PDC construct can be improved through the use of nanomaterials.
`5. Bicycle-toxin conjugates (BTCs) and peptide-dendrimer conjugates are emerging constructs that fall under the umbrella term PDCs.
`
`Introduction
`
`Traditionally, research within drug discovery falls into two
`groups: small molecules (o500 Da) and biologics (45000 Da).1
`Peptides are placed within the molecular weight range that is
`typically under-represented in the pharmaceutical company
`pipelines. A peptide is defined by the FDA as a polymer
`composed of less than 40 amino acids (500–5000 Da).2 Over
`recent years, the research community is acknowledging the
`many advantages that peptides bring over small molecules and
`biologics. These include simpler design, ability to interact with
`underexplored targets, cheaper synthesis, decreased immuno-
`genicity and enhanced tissue penetration.1 To date in the U.S.,
`
`a Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge,
`CB2 1EW, UK. E-mail: spring@ch.cam.ac.uk
`b Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, UK
`c Medicinal Chemistry, Research and Early Development Cardiovascular,
`Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg,
`Sweden
`
`Europe and Japan markets, there are more than 100 peptide
`drugs used to treat a range of diseases.2 Financially, the peptide
`market is lucrative as it is estimated to be worth d11–16 billion
`annually by 2019.2 However, there is still a significant challenge
`for the pharmaceutical industry to get peptides to market with
`many adopting greener peptide synthesis techniques at
`increased costs than traditional approaches.
`Peptides can offer a multifunctional approach – in addition
`to being biologically active, they are excellent at transporting
`cargos to the desired targets. Their use within targeted ther-
`apeutics is an exciting area of research with great promise in
`the future with particular focus in, but not limited to, oncology.
`Witnessing the current success and investment into many
`antibody–drug conjugates (ADCs), the equivalent peptide–drug
`conjugates (PDCs) show promise for the future of the use of
`peptides within this setting. This review will highlight the
`excellence and limitations of peptides, their use in PDCs for
`advancing targeted cancer therapeutics and will consider how
`the specific tumour microenvironment can aid the design of a
`PDC. In addition, the review provides an examination of how
`
`1480 | Chem. Soc. Rev., 2021,50 , 14801494
`
`This journal is
`
`The Royal Society of Chemistry 2021
`
`View Article Online
`
`View Journal
`
` | View Issue
`
`MPI EXHIBIT 1071 PAGE 1
`
`MPI EXHIBIT 1071 PAGE 1
`
`

`

`Tutorial Review
`
`Chem Soc Rev
`
`the stability of PDCs in circulation can be enhanced through
`both chemical modifications and material science – a
`topic rarely discussed but extremely valuable to the successful
`development of new peptide drugs.
`
`ADME & PK considerations for peptides
`
`When considering therapeutic agents, it is crucial to analyse
`pharmacokinetic (PK) properties such as absorption, distri-
`bution, metabolism and excretion (ADME) as well as pharma-
`codynamic (PD) properties. Traditionally, PK properties of
`peptides tend to differ substantially from those of small drug
`molecules.3 A significant limitation of peptides is their limited
`or non-existent oral bioavailability leading to administration
`
`through intravenous (IV) injection. One of the most convenient
`methods of administration is oral as it allows the patient to
`take the medication independently, unlike IV that often
`requires a clinical setting.
`A small molecule is defined as ‘drug-like’ if it satisfies the
`criteria of Lipinski’s Rule of 5 (Ro5). These rules focus on
`several fundamental factors including the molecular weight
`(o500 Da), r5 H-bond donors, r10 H-bond acceptors and
`log P (o5): molecules that satisfy the Ro5 are likely to be orally
`bioavailable.4 Peptides do not fit into the Ro5 criteria due to
`their relatively large size (500–5000 Da) in comparison to small
`molecules (typically o500 Da). Despite peptides not satisfying
`the Ro5 criteria, the rule does not indicate that a peptide
`cannot become a drug as proven through many peptides on
`the market and in clinical trials.4
`
`Bethany M. Cooper received her
`MChem degree from the Univer-
`sity of Leeds in 2018, having
`completed her 3rd year of study
`at Lubrizol Ltd, Hazelwood. On
`return to the University of Leeds
`her final year was spent under the
`supervision of Professor Steve
`Marsden. In 2019, she started
`her PhD studies at the University
`of Cambridge under the super-
`vision of Professor David Spring
`and industrial
`supervisor Dr
`Maria O¨lwegård-Halvarsson, where
`her research has focused on peptide stapling methodologies for use
`within therapeutics.
`
`Bethany M. Cooper
`
`Jessica Iegre was born in Italy
`and
`obtained
`her MSci
`in
`Medicinal Chemistry and Pharma-
`ceutical
`Technology
`at
`the
`University of Pisa, Italy in 2013.
`The same year, she joined the
`AstraZeneca IMED Graduate pro-
`in Go¨teborg, Sweden
`gramme
`where she spent 2 years working
`across three different departments:
`medicinal chemistry, DMPK and
`computational chemistry. In 2015
`she joined the Spring group at the
`University of Cambridge, and she
`obtained her PhD in chemical biology in 2019. Jessica is currently a
`Postdoctoral Research Associate in the group, and she is developing
`novel stapled peptides to inhibit medicinally relevant protein–protein
`interactions.
`
`Jessica Iegre
`
`Daniel Hillebrand O’ Donovan is
`currently
`Associate
`Principal
`Scientist in Early Oncology R&D
`at AstraZeneca, Cambridge, UK.
`Following PhD studies in medi-
`cinal chemistry at Trinity College
`Dublin, he moved to Germany for
`postdoctoral studies at the Max
`Planck
`Institute
`for Kohlen-
`forschung followed by a Marie
`Curie
`fellowship at
`the Uni-
`versity of Oxford, UK. Since
`joining AstraZeneca in 2016, he
`has worked in a variety of
`including antihormonal
`therapies
`for breast
`research areas
`cancer,
`epigenetics,
`immuno-oncology and targeted protein
`degradation.
`
`Daniel H. O’Donovan
`
`Dr Maria O¨lwegård-Halvarsson is
`a Senior Research Scientist in the
`New Modalities group, Depart-
`ment of Medicinal Chemistry,
`AstraZeneca, Sweden.
`In her
`current role she develops linker
`chemistry and synthesizes drug
`conjugates within the targeting
`delivery platform. She obtained
`her PhD in organic chemistry
`from Gothenburg University
`in
`1990 and then spent four years at
`the Department
`of Medicinal
`Biochemistry, Gothenburg Uni-
`versity. She joined AstraZeneca in 1995, working three years in the
`isotope labelling group and has a 25 year commitment to the company,
`synthesizing drug molecules within the cardiovascular and metabolic
`disease areas in lead optimization and lead generation phase.
`
`Maria O¨lwegård Halvarsson
`
`This journal is
`
`The Royal Society of Chemistry 2021
`
`Chem. Soc. Rev., 2021,50 , 14801494 | 1481
`
`View Article Online
`
`MPI EXHIBIT 1071 PAGE 2
`
`MPI EXHIBIT 1071 PAGE 2
`
`

`

`Chem Soc Rev
`
`Tutorial Review
`
`enzymes that are responsible for the chemical instability and
`the breakdown of peptides in the bloodstream.
`Short half-lives are also experienced due to rapid renal
`clearance, resulting in many hampered peptide in vivo studies
`and ultimately the pursuit of the peptide as a drug. Found
`within the kidney are glomeruli pores that have a size of
`B8 nm; circulating peptides that are less than 25 kDa filter
`through the glomeruli, and are not reabsorbed through the
`renal tubule.3 Considering such limitations, it is not surprising
`that many oral peptide drug candidates have entered clinical
`trials, but with limited success and overall are restricted to the
`area of endocrine disorders. Formulations are helpful in this
`setting and have been successfully used in the development of
`Semaglutide, the most recently FDA approved orally available
`peptide drug used to treat Type 2 diabetes (September 2019).
`Sodium N-[8-(2-hydroxybenzoyl)amino caprylate]
`(SNAC)
`is
`used with Semaglutide to form a co-formulation.7 SNAC works
`as a buffering agent within the stomach, which in turn
`diminishes the activity of proteolyzing enzymes including
`pepsin where the maximum activity is experienced at a pH
`within the range of 2–4.7 Despite Semaglutide’s approval, the
`delivery of peptides orally still has a long way to develop before
`we see more in the clinic.
`Several ways can be used to try and improve the ADME
`properties of peptides such as the improvement of the cell
`permeability, enhancement of chemical and proteolysis stability
`and reduction of renal clearance overall resulting in the
`extension of the circulatory half-life. The extended half-life is
`beneficial both economically and for the patients’ compliance.
`The next part of the review will focus on the ways of modifying a
`peptide to achieve such improvements.
`
`Improving enzymatic and chemical
`stability of peptides by chemical
`modifications
`Cyclisation
`
`Cyclisation techniques have been used widely in the peptide
`field and achieved in several ways from cyclising head to tail,
`head/tail to side chain or side chain to side chain.
`A type of side chain to side chain cyclisation is called
`stapling, a technique that enables the peptide to be locked into
`a desired conformation. Peptide stapling is used commonly to
`enhance a peptide’s secondary structure such as a-helices and
`b-turns, which can improve the binding affinity to the target
`and enhance ADME properties.
`There are two subgroups of peptide stapling (PS): one-
`component (1C) and two-component (2C). In one-component
`peptide stapling (1C-PS) there is an intramolecular linkage
`between often unnatural amino acid side chains and can allow
`cyclisation depending on the secondary structure.
`One of the first examples of 1C-PS was by Blackwell and
`Grubbs through the use of ring-closing metathesis (RCM) of O-
`allyl serine residues.8 1C-PS is not without its limitations. For
`example, modifications would result in the entire peptide being
`
`Fig. 1 A schematic to represent the journey of an orally administrated
`peptide through the gastrointestinal tract.
`
`Santos et al. analysed peptides approved by the FDA between
`2012–2016 to allow comparison to the Ro5.4 They determined
`that the most orally available peptides were indeed up to a MW
`of 1200 Da and displayed a log P within the range of 5–8. Close
`interpretation of the results found that orally available peptides
`had 5 times more H-bond donors and acceptors than what was
`considered as acceptable by Lipinski’s Ro5 for small molecules.4
`Oral administration is challenging for both biologics and
`peptides and in many cases, it may not be feasible. The whole
`journey of oral administration through the gastrointestinal
`tract is problematic, starting with the enzymes amylase and
`lipase found within saliva that break down the peptides into
`smaller molecules (Fig. 1).5 On arrival into the stomach, the
`peptide is subjected to harsh acidic conditions and proteolysis
`by cathepsin and pepsin. Even if the peptide successfully
`remains intact to this point, the lumen of the small intestine
`experiences a pH change and has a vast number of proteolyzing
`enzymes including trypsin, chymotrypsin and carboxypeptidase.5
`Compared to biologics, peptides have a much shorter circu-
`latory half-life (days vs. weeks) resulting in the need for sub-
`optimal frequent drug administrations. The impact of this is
`witnessed through the advancement of ADCs and a slower
`progression of PDCs. The lifetime of a hydrophilic peptide in
`circulation is determined by many soluble enzymes in the
`blood and at membranes. Exopeptidases are a class of enzymes
`that can be split into two subgroups: amino- and carboxypepti-
`dases and target the N- and C-terminal, respectively.6 It is these
`
`currently
`is
`Spring
`David
`of Chemistry
`and
`Professor
`Biology
`at
`the
`Chemical
`University of Cambridge within
`the Chemistry Department. He
`received his DPhil
`(1998) at
`Oxford University under Sir Jack
`Baldwin. He then worked as a
`Wellcome
`Trust
`Postdoctoral
`Fellow at Harvard University
`with Stuart Schreiber
`(1999–
`2001), after which he joined the
`faculty at the University of Cam-
`bridge. His research programme
`is focused on the use of chemistry to explore biology.
`
`David R. Spring
`
`1482 | Chem. Soc. Rev., 2021,50 , 14801494
`
`This journal is
`
`The Royal Society of Chemistry 2021
`
`View Article Online
`
`MPI EXHIBIT 1071 PAGE 3
`
`MPI EXHIBIT 1071 PAGE 3
`
`

`

`Tutorial Review
`
`Chem Soc Rev
`
`resynthesized, which could prove costly in both time and
`materials. However, 1C-PS has proved successful in many cases
`exemplified by the first stapled peptide ALRN-5281 to reach
`clinical trials and complete Phase I.9 ALRN-5281 is used to treat
`adult growth hormone deficiency and was cyclised through the
`use of 1C-PS via RCM.9
`Peptide stapling more recently has moved the focus away
`from 1C-PS to two-component peptide stapling (2C-PS) (Fig. 2).
`The use of 2C-PS offers many advantages over 1C-PS and allows
`synthetic versatility through the use of linear peptides and
`separate staples. 2C-PS allows modifications to be made at a
`late stage if needed to the peptide or the staple, which is highly
`beneficial for an optimization campaign. 2C-PS has been widely
`applied to bicycles therapeutics developed by Christian Heinis
`and Sir Greg Winter.10 The overall concept of bicycles is the
`cross-linking of three cysteine residues to a tri-functionalised
`linker to form a bicycle construct. More details on the application
`of bicycles for use within drug conjugates are detailed later in
`the review.
`
`Moving away from proteinogenic amino acids
`
`Side chains of amino acids offer another excellent source for
`modification with many papers being published recently on
`direct amino acid modification. Increasing the steric bulk of
`the side chains results in increased stability, as enzyme recog-
`nition is disrupted.6
`One way to increase the overall stability of the full peptide is
`to swap L-amino acids to D-amino acids. The D-amino acid
`sequence has a decreased substrate recognition and binding
`
`affinity for proteolytic enzymes.3 An example of increasing the
`half-life of a biologically active peptide is modifying Somatos-
`tatin to Octreotide used to treat gastrointestinal tumours.3,6
`Octreotide’s amino acid sequence incorporates two D-amino
`acids, whereas Somatostatin is only composed of L-amino acids
`(Fig. 2). The resulting half-life increases from a few minutes for
`Somatostatin to 1.5 hours for Octreotide hence enhancing
`favourable PK properties.11 Despite this example highlighting
`the benefits of L- to D-amino acid exchange, there are a few
`cases in which D-amino acid-containing peptides show a
`reduced half-life in comparison to the L-analogue.3 It is important
`to consider the effects that such modifications could have on
`the overall secondary structure of the peptide and on any intra-
`or intermolecular interactions.3 An alternative is the use of
`D-peptides. These peptides are mirror images of the L-amino
`acid-containing counterpart and are composed fully of D-amino
`acids. The Kay group have recently developed D-peptides for use
`as HIV entry inhibitors.12
`
`Slowing down renal clearance by chemical modifications
`
`The overall net charge for the peptide sequence is an important
`consideration for renal clearance. Peptides that acquire a net
`negative charge tend to exhibit a longer half-life in comparison
`to those with a net positive charge.13 The presence of anionic
`carbohydrate moieties found within the kidney’s glomerular
`membrane limit the filtration of anionically charged species
`into the urine.13
`Another approach is the conjugation of peptides with larger
`molecules (450 kDa) to increase lipophilicity and their binding
`
`Fig. 2 Different methods used to improve the enzymatic and chemical stability of peptides including: cyclisations and peptide stapling, formulations,
`moving away from proteinogenic amino acids and chemical modifications.
`
`This journal is
`
`The Royal Society of Chemistry 2021
`
`Chem. Soc. Rev., 2021,50 , 14801494 | 1483
`
`View Article Online
`
`MPI EXHIBIT 1071 PAGE 4
`
`MPI EXHIBIT 1071 PAGE 4
`
`

`

`Chem Soc Rev
`
`Tutorial Review
`
`to albumin, thereby improving the PK and PD properties of
`peptides.13 It is the enhanced steric that prevent the conjugate
`from being filtered out through the kidneys and allow a longer
`circulation period. Modifications at the N- and C-terminus help
`slow down renal clearance. Typically the breakdown by exopepti-
`dases of a peptide sequence occurs at the N- or C-terminus.6
`There are ways this can be prevented by modifying the
`terminus to increase proteolysis resistance. An amide bond
`between the C- and N-terminus achieved through a cyclisation
`reaction has been shown to prevent enzyme degradation.6
`If cyclisation is not the preferred structure for binding, then
`N-acetylation and C-amidation can be an alternative to enhance
`resistance to proteolysis. These modifications have proven
`successful in a range of studies on Somatostatin, where the
`half-life of the modified molecule was extended when com-
`pared to the unmodified peptide.3
`N-Methylation of amide bonds is another modification
`that enhances metabolic resistance. N-Methylation increases
`the steric hindrance and allows tuning of
`the peptide
`conformation.14 Cyclosporin is an example of a naturally occur-
`ring peptide used as an immunosuppressive drug (Fig. 2).
`Cyclosporin is a hepta-N-methylated cyclic undecapeptide, with
`an oral bioavailability of 29%.14
`(PEG) has been vastly
`The use of polyethylene glycol
`explored in slowing down renal clearance and by increasing
`the binding to plasma proteins such as albumins. PEG is an
`ideal candidate for modification: it is cheap, biocompatible,
`hydrophilic, non-toxic and non-immunogenic.6 The promise of
`PEGylation for the modification of peptides is highlighted by a
`number of examples, which are discussed herein.
`RGD is a homing tripeptide (see later), whose sequence is
`able to allow the HM-3 peptide to selectively bind to specific
`target sites that display high levels of
`integrins within
`tumours.13,15 The original HM-3 peptide had limited effect as
`a consequence of its short half-life and required twice a day
`administration. There was a need to enhance the peptide’s
`half-life to reduce the number of administrations needed.
`Methoxy-poly(ethylene glycol)-aldehyde (mPEG-ald) was the PEG
`linker of choice for attachment at the N-terminus. Upon this
`modification, there was a 5.86 fold increase in half-life in male rat
`studies when compared to the unmodified HM-3 peptide.15
`Another PEGylated peptide is PEG-adrenomedullin (PEG-ADM)
`by Bayer used in patients suffering from Acute Respiratory
`Distress Syndrome (ARDS) associated with lung failure.16 The
`peptide was enrolled into Phase 2 clinical trials in August 2020
`with the predicted end date of early 2023.16
`PEG is not the only molecule used in conjugation to the
`peptide approaches to slow down renal clearance. Other widely
`used examples include polysialic acids (PSA), a homopolymer,
`and hydroxyethyl starch (HES), a branched amylopectin.13
`The addition of fatty chains has been an effective method as
`an addition to peptides to increase the half-life. Glucagon-like
`peptide (GLP-1) receptor agonists have been used to control
`blood sugar levels in patients with Type 2 diabetes.17 One of the
`earliest example is Exenatide, an analogue of a nonhuman
`peptide that in 2005 had twice-daily administration and an IV
`
`Fig. 3 The structures of Liraglutide and Semaglutide.
`
`half-life of 30 minutes. In 2009 Liraglutide, a near analogue of
`human GLP-1, had a fatty acid chain with a spacer joined to
`the main peptide backbone for binding to albumin (Fig. 3).
`Liraglutide represented a significant improvement of GLP-1
`with an extended IV half-life of 8–10 hours and once-daily
`administration.17 Semaglutide is a GLP-1 agonist, with a
`gGlu-2xOEG linker to a C18 fatty chain (Fig. 3).17 The determined
`IV half-life was 46.1 hours through studies involving mini pigs,
`enabling once-weekly doses – a vast improvement to the early
`GLP-1 agonists.17
`
`Enhancing bioavailability via formulations
`
`Several methods have been used previously in the literature to
`enhance the oral bioavailability of peptide therapeutics via
`formulations.5 These can include permeation enhancers and
`acid-stable coatings (Fig. 2).5 Permeation enhancers are able to
`transport the peptide through epithelial cells – an alternative
`route is available through intercellular junction and adhesion
`protein interference resulting in a paracellular route.5 The use
`of acid-stable coatings to improve the oral availability of a drug
`is a widely used approach. These coatings are pH active, where
`at low pH in the stomach the coating remains intact; as the
`peptide moves to the intestine the pH rises and the coating
`breaks down to release the contents. The introduction of citric
`acid can be used to help neutralise the optimum basic pH
`conditions for a range of gastrointestinal peptidases, hence
`slowing down degradation caused by peptidases.5
`Formulations can be used to enhance the bioavailability of
`IV administrated peptides. The FDA approved Sandostatin LAR
`is an excellent example of this in which Octreotide is encapsulated
`in a glucose-poly(lactide-co-glycolide) (Glu-PLGA) star-shaped poly-
`mer (Fig. 2).18
`Improving the overall chemical and enzymatic stability of a
`peptide using the techniques discussed is beneficial for the
`discovery of new therapeutics including targeted therapies
`exemplified by peptide–drug conjugates. These concepts will
`be discussed in the following section.
`
`Cancer therapy and targeted drug
`delivery
`
`Cancer is among the leading contributors to human mortality
`and disease, with nearly 50% of people being diagnosed with
`
`1484 | Chem. Soc. Rev., 2021,50 , 14801494
`
`This journal is
`
`The Royal Society of Chemistry 2021
`
`View Article Online
`
`MPI EXHIBIT 1071 PAGE 5
`
`MPI EXHIBIT 1071 PAGE 5
`
`

`

`Tutorial Review
`
`Chem Soc Rev
`
`cancer in their lifetime.19 In 2000 and 2010, two landmark
`publications defined the ‘‘Hallmarks of Cancer’’, characterising
`all cancers with eight common traits: cancer cells stimulate
`their own growth, lack sensitivity to anti-growth signals, evade
`programmed cell death (apoptosis), can divide indefinitely, can
`sustain blood vessel formation (angiogenesis), invade other
`tissues and metastasise, deregulate metabolism and evade the
`immune system.20,21 Our growing understanding of cancer cell
`biology has enabled significant advances in treating this disease.
`Subject to the stage and tumour type, patients are treated
`with one or a combination of the following options: surgery,
`radiotherapy or pharmacotherapy.
`is charac-
`Traditional pharmacotherapy (chemotherapy)
`terised by cytotoxic drug regimens which target rapidly dividing
`cells through inhibiting mitosis and is associated with serious
`side-effects such as bone marrow and gastrointestinal toxicity.
`Even if the tumour is successfully eradicated, lasting damage
`may continue to affect healthy tissues and residual cancer cells
`may result in relapse of the disease.
`Fortunately, newer molecular therapies can improve patient
`outcomes and reduce toxicity by targeting the unique charac-
`teristics of tumour cells including the cell morphology (leaky
`cell membranes), lower pH, increased glutathione (GSH) and
`enzyme presence (Fig. 4).22
`Antihormonal therapy is often effective for tumours whose
`growth is driven by endocrine signalling, such as breast and
`prostate cancers. Drugs which selectively inhibit an oncogenic
`mutant protein while sparing the unchanged (wild-type) protein
`are another successful approach, such as Gleevec (imatinib), a
`tyrosine kinase inhibitor which has revolutionised the treatment
`of Chronic Myelogenous Leukemia (CML).23 By targeting cancer’s
`ability to evade the immune response, checkpoint inhibitor
`antibodies such as Keytruda (pembrolizumab) and Imfinzi
`(durvalumab) can rekindle the immune system’s ability to recog-
`nise and eliminate tumours, providing new treatment options for
`recalcitrant tumours such as non-small lung cell cancer (NSCLC)
`and Hodgkin’s lymphoma. Following treatment with all of these
`
`Fig. 4 A schematic representation of how a targeted therapeutic (green
`square) is specific for the tumour cells in comparison to a non-targeted
`therapeutic (blue triangle).
`
`drugs, cancers may further mutate to develop resistance to
`targeted therapies; these resistance mutations provide challenges
`but also new opportunities for drug discovery.24
`Antibody–drug conjugates (ADCs) are another promising
`line of targeted therapy composed of an antibody which binds
`to a protein highly expressed in tumour cells (a tumour antigen),
`connected via a linker to a cytotoxic small molecule payload.
`In principle, this design enables the targeted delivery of a drug
`to the tumour, killing cancer cells while sparing healthy tissue
`and providing a wider therapeutic margin than traditional
`chemotherapy. In 2000, Mylotarg (gemtuzumab ozogamicin)
`became the first ADC approved by the FDA, combining an
`antibody to the CD33 antigen expressed in leukaemia cells with
`a cytotoxic payload derived from a calicheamicin natural product.
`Over the past 20 years, there have been significant advances in
`this field exemplified by 5 ADCs approved between 2008 to 2018
`and 3 ADCs approved by the FDA in 2019 for various cancers,
`including AstraZeneca and Daiichi’s Enhertu (trastuzumab der-
`uxtecan) for the treatment of HER2 metastatic breast cancer,
`Roche’s Polivy (polatuzumab vedotin) for large B-cell lymphoma,
`and Astella and Seattle Genetics’ Padcev (enfortumab vedotin-ejfv)
`for urothelial bladder cancer.25 The most recent ADC being
`approved in August 2020 was GSK’s Blenrep (belantamab
`mafodotin-blmf) for relapsed or refractory multiple myeloma,
`bring the total to 9 FDA approved ADCs as of September 2020.26
`Despite continuing progress for ADCs, these therapies pre-
`sent several drawbacks. As cytotoxic drugs, ADCs can incur
`significant toxicity; in the case of Mylotarg, the FDA issued a
`black box warning for potentially life-threatening side effects in
`patients who did not receive stem cell transplantation.27 ADCs
`can also generate dangerous immune reactions, yielding toxi-
`city and halting further therapy.28 Furthermore, the compli-
`cated structure of ADCs results in high production costs, in
`some cases restricting access to these drugs as their cost-
`effectiveness may be called into question by health insurers.
`Owing to their high molecular weight and protein-like physico-
`chemical properties, ADCs also suffer from restricted distri-
`bution which can prevent these therapies from penetrating some
`types of solid tumours and limiting their efficacy.
`To overcome these issues, many research groups have
`explored alternative approaches such as peptide-based drugs,
`protein–protein interaction inhibitors (PPIs) and drug delivery
`transporters. This review will focus on the latter and describe
`how peptides can be used within peptide–drug conjugates
`(PDCs) for the targeted treatment of cancer (Fig. 5).
`
`Peptide–drug conjugates (PDCs)
`
`Peptide–drug conjugates are a class of targeted therapeutics,
`with a similar construct to that of ADCs and only differing
`through the homing device. A PDC is composed of three vital
`components: a homing peptide, a linker and a cytotoxic pay-
`load (Fig. 5). All three work in synergy to deliver cytotoxins
`through targeting the selected receptor of a tumour cell. As
`discussed earlier, the ADC market has been fast-paced, but
`
`This journal is
`
`The Royal Society of Chemistry 2021
`
`Chem. Soc. Rev., 2021,50 , 14801494 | 1485
`
`View Article Online
`
`MPI EXHIBIT 1071 PAGE 6
`
`MPI EXHIBIT 1071 PAGE 6
`
`

`

`Chem Soc Rev
`
`Tutorial Review
`
`These PDCs are examples of therapeutics, however, PDCs
`can be used as successful diagnostic tools by employing the use
`of radionucleotides.
`On constructing a PDC, it is key to understand the role of
`each component to fully appreciate the design. Considerations
`include the mechanism of action and how alternative
`approaches can improve the current limitations of this emer-
`ging modality.
`
`Homing peptide
`
`A homing peptide is a selected peptide that is chosen for its
`specific targeting capabilities of protein receptors found over-
`expressed at tumour tissues. In the case of PDCs, the homing
`peptide will direct the whole PDC construct to the targeted cell
`and limit off-target delivery. These homing peptides often have
`precedent in the literature for having a strong binding affinity
`to the target site within the nanomole magnitude. Several
`techniques can be used to determine their binding affinity
`including surface plasmon resonance (SPR), biolayer interfero-
`metry (BLI) and isothermal titration calorimetry (ITC).
`The secondary structure of the homing peptide has a pro-
`nounced effect on its binding affinity. Therefore, structural
`information is important when seeking to increase the binding
`affinity of the homing peptide through stabilisation of the
`secondary structure. The most common examples of peptide
`secondary structure include a-helix, b-sheet and random coil.
`On the attachment of the linker to allow conjugation to the
`cytotoxin, the secondary structure of the homing peptide must
`be retained, and the linker should not disrupt binding. There is
`a vast range of homing peptides for many targets detailed in a
`recent review by Vrettos et al. and summarised in Table 1.19
`Most homing peptides reported to date are linear. Even
`though they show good binding, there are several drawbacks
`including degradation by enzymes at the termini, chemical
`instability and fast renal-clearance. A way to overcome these
`limitations is through the use of cyclisation or peptide stapling
`of the linear peptide as described earlier.
`A study by Lu and co-workers demonstrated how a stapled
`RGD peptide was used as a homing peptide that targets avb3
`integrin (Fig. 6).31 The peptide can be used to modify a
`nanoparticle to effectively deliver a drug to target glioblastoma
`multiforme (GBM), an aggressive CNS tumour with poor
`prognosis.31 The RGD tripeptide has precedent for targeting
`avb3 integrins expressed on glioma cells and overcoming the
`blood brain tumour barrier (BBTB). During the early stages of
`glioma, therapeutics can be hindered as the blood brain barrier
`(BBB) still remains intact. Therefore, a suitable vehicle to
`
`Fig. 5 A schematic of a peptide–drug conjugate construct consisting of a
`homing peptide, linker and payload. The structure of 177Lu-dotatate an
`FDA approved peptide–drug conjugate.
`
`unfortunately, this is not the same case for PDCs. A contributing
`factor is that methodologies that overcome the intrinsic poor PK
`properties of the peptides have only recently emerged, and
`pharmaceutical companies have started expanding their pipelines
`to accommodate peptide therapeutics only in the last decade.
`There is currently only one therapeutic PDC on the market,
`177Lu-dotatate, but many more are in various phases of the
`pipeline. 177Lu-dotatate is used to treat gastroenteropancreatic
`neuroendocrine tumours (GEP-NETs) and was the first FDA
`approved PDC (Fig.

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket