<<

DRUG IN Clinical Medicine 2017 Vol 17, No 3: 220–232

Developments in with monoclonal and related proteins

Authors: H M i c h a e l S h e p a r d ,A G a i l L e w i s P h i l l i p s ,B C h r i s t o p h e r D T h a n o sC a n d M a r c F e l d m a n nD

Monoclonal therapeutics have been approved will focus on mAbs and relatives, describing these therapeutics. for over 30 targets and , most commonly cancer. The tumour necrosis factor (TNF)-blockers for autoimmune/ Antibodies have become the new backbone of the inflammatory diseases are the most broadly deployed pharmaceutical industry, which previously relied on small (with multiple products) and have engendered a revolution molecules. Compared with small molecules, monoclonal in therapeutic research/development, along with rather ABSTRACT antibodies (mAbs) have exquisite target selectivity and hence remarkable revenues. This therapeutic revolution is based less toxicity as a result of binding other targets. The clinical on the synergy of three scientific disciplines: immunology, value of both mAbs and ligand traps has been proven. New molecular biology and protein engineering. applications of mAbs are being tested and mAbs have now been designed to target two (bi-specific, eg TNF-α and IL-17) B r i e f h i s t o r y or more targets simultaneously, augmenting their therapeutic potential. Because of space limitations and the wide ranging The first mAbs were produced by Milstein and Köhler 1 scope of this review there are regrettably, but inevitably, (Cambridge University), who won the Nobel Prize in omissions and missing citations. We have chosen to highlight Physiology or Medicine in 1984. mAbs were initially murine the first successes in inflammatory diseases and cancer, but a proteins and hence were immunogenic in humans and broader overview of approved mAbs and related molecules can not suited for chronic therapy. This limitation was solved be found in Table 1. using molecular biology and protein engineering to create more human-like mAbs with little immunogenicity. There K E Y W O R D S : Biologic therapy , cancer , infl ammation , monoclonal are several versions of mAbs and traps in clinical use and antibodies , traps trials (Fig 1 , Table 1 ). Initial attempts replaced most murine Fc sequences with human Fc, a process referred to as ‘chimerisation’.2 This involves grafting the murine Introduction binding Fab regions onto a human immunoglobulin (IgG) backbone. In ‘humanisation’, the mouse hyper-variable There has been a revolution in therapeutics over the past peptide binding loops 3 are grafted onto the human IgG 20 years, due to the introduction of protein therapeutics – backbone. More recent technologies allow for generation of often termed ‘biologics’ – into routine, mainstream medicine. fully human antibodies.4 Biologics are expensive and like all medicines have risks. Their advantage is exquisite specificity due to greater surface area binding, resulting in decreased ‘off-target’ effects as compared Improved therapy for inflammatory/autoimmune with most small molecule drugs. New biologic therapies come in several basic forms, either The concept of autoimmune diseases, pioneered by Sir Frank growth factors and (such as , G-CSF, McFarlane Burnet 5 (Nobel Laureate 1960), was validated by interferon, enzymes, factors that regulate coagulation) or, more the detection of serum autoantibodies in many diseases in the commonly, monoclonal antibodies (mAbs) and related proteins 1960s. This suggested a role for B-lymphocytes, and human such as ‘traps’ in which receptors are made soluble leukocyte antigen (HLA) associations documented from the and fused with antibody constant regions. The latter group 1970s indicated the role of T-cells when the peptide presenting (mAbs and traps) have dramatically advanced the therapy of function of HLA was understood. However, these advances did chronic inflammatory diseases and cancer. In this review, we not lead to new treatments at the time because of the lack of specific therapeutics. With the advent of mAbs, the role of T-cells was tested in Authors: Aresearch fellow, Halozyme, San Diego, USA; B senior rheumatoid arthritis patients using lytic antibodies to CD4, scientist, Inc, San Francisco, USA; Csenior director, without clinical benefit.6 Nor did small trials of several other New Molecule Discovery, Halozyme, San Diego, USA ; Demeritus anti-T-cell antibodies, such as anti-CD7. OKT3, a mouse mAb professor, Kennedy Institute of Rheumatology, Oxford, UK recognising CD3 7 given to reduce acute rejection in patients

220 © Royal College of Physicians 2017. All rights reserved.

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 220220 222/05/172/05/17 9:479:47 PMPM Monoclonal antibodies

ABCAngen (eg cancer target) Angen within Angen-binding site angen-binding site Heavy (H) V chain Murine Chimeric Humanised Fully human ADC structures H V H VL VL

C 1 CH1 Light (L) H CL CL chain Hyper-variable regions -omab -ximab -zumab -umab Lys Cys Site-specific Disulphide CH2CH2 bonds HighPotenal for immunogenicity Low Carbohydrate Effector region

CH3 CH3

Fig 1. Monoclonal antibodies. A – antibody structure showing light (L) and heavy (H) chains with variable (VL , VH ) and constant (CL , CH ) regions, which are connected by inter-chain disulphide bonds. Antigen binding (green symbol) occurs at VH /VL domains, while effector functions are mediated via the Fc (CH ) portion. B – increasing the amount of human sequences in a murine antibody decreases immunogenic potential. C – on an antibody-drug conjugate (ADC, cy- totoxic drugs (orange, red stars) can be linked stochastically to lysine (Lys) residues or on cysteine residues either through reduction of inter-chain disulphides (Cys) or by engineering in cysteines at select conjugation sites (site-specifi c ).

with organ transplants was the first of methotrexate,12 and some patients can be taken off all approved for use in humans although it was later withdrawn. .13 In the wake of anti-TNF's success, other mAbs have also The success of anti-TNF therapy proven useful in inflammatory diseases. The antibody to the 14 The targeting of a single pro-inflammatory cytokine, TNF, to IL-6 receptor, tocilizumab (Actemra®), pioneered by Professor treat a complex disease in rheumatoid arthritis (RA) where Tadamitsu Kishimoto, has been approved for RA and for multiple pro-inflammatory cytokines were upregulated was juvenile idiopathic arthritis. Currently, other anti-cytokine based on work using human disease tissue by Feldmann antibodies to IL-6, GM-CSF and the GM-CSF receptor have and Maini8 and Brennan et al.9 They analysed cytokine been approved ( (Sylvant®) – anti-IL-6) or are in late stage clinical trials after successful phase II studies. production from joints and cytokine regulation in cultures 15 of rheumatoid synovium in which the majority of the cells , a chimeric mAb to CD20, (an antigen in most survived, producing the mediators generated in vivo . In these B-cells although not plasma cells), was first to treat B-cell- cultures, blocking TNF-α reduced the production of many driven cancers such as non-Hodgkin . It was other inflammatory cytokines (IL-1, IL-6, GM-CSF, IL-8 pioneered by Jo Edwards for RA and subsequently approved, etc),9 thus defining a ‘TNF-dependent cytokine cascade’. The but was not successful in systemic lupus erythematosus dramatic clinical success of TNF blockade, demonstrated first (SLE) trials. Anti-CD52 () is a first generation in late stage RA then in earlier stage disease, also validated humanised antibody, now used in multiple sclerosis. There 8 are other antibodies approved, eg belimumab (also known as this concept. Noteworthy was the fact that tissue ( 16 and cartilage) damage was controlled. But also striking Benlysta) is an anti-BLys mAb approved for SLE, u s t e k i n u m a b was the heterogeneity of the clinical response, with some (also known as Stelara) is an antibody to the shared p40 subunit of IL-12 and IL-23 approved for psoriasis and psoriatic individuals close to a cure and others virtually unimproved. 17 The reasons for this are not yet clear, despite much work to try arthritis and potentially for Crohn's disease, and secukinamab (Cosentyx) is an anti-IL17A mAb approved for severe psoriasis to elucidate the reasons. Genetic differences were an obvious 18 possibility although never established, and recent clinical data and ankylosing spondylitis. demonstrating that non-responders may respond subsequently Receptor IgG fusion proteins (‘ligand traps’) have also been generated. TNF receptor 2-Fc (etanercept, Enbrel®) is to anti-TNF has excluded it. Although anti-TNF in humans 19 is relatively safe,10 more in patients occur, eg with effective and has been approved. Other ligand traps include intracellular organisms, especially tuberculosis. Many large rilonacept (Arcalyst – an IL-1 Trap) and the anti- patient registers have documented the long-term benefits vascular endothelial trap (aflibercept (Eylea®) of anti-TNF therapy, reducing some complications and for retinopathies and ziv-aflibercept (Zaltrap®) for colorectal maintaining a favourable benefit/risk ratio.11 cancer) (Table 1). Abatacept (Orencia®) and belatacept Currently, anti-TNF is used in RA, Crohn's disease, ulcerative (Nolojix®) are CTLA4-Fc fusion proteins that inhibit antigen colitis, psoriasis, psoriatic arthritis, ankylosing spondylitis and presentation to T-cells and are effective in RA. juvenile RA; its use is now being explored in other indications (Table 1) . Anti-TNF antibodies are the most successful and Monoclonal antibodies in cancer therapy widely used antibody-based therapeutic. It is noteworthy mAb discovery: a case study of (Herceptin) that if used together with methotrexate early in the course of RA, over 50%12 of patients can be taken off infliximab Modern mAb therapy of solid tumours was initiated by the and remain virtually disease-free, even with reduced dosage humanised human epidermal 2 (HER2)

© Royal College of Physicians 2017. All rights reserved. 221

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 221221 222/05/172/05/17 9:479:47 PMPM H Michael Shepard, Gail Lewis Phillips, Christopher D Thanos et al Mouse/human IgG1 fragment Fab Trade name changed from Campath- name changed from Trade (multiple to Lemtrada 1H (cancer) sclerosis) Humanised IgG1k Basiliximab: chimeric mouse/human IgG1k. glycoengineered IgG1 mAb with glycoengineered enhanced ADCC. Rituximab is chimeric mouse/human IgG1k and tositumumab are radio-conjugates be used when tumours stop that can responding to the anti-CD20 mAbs. Ibritumumab and tositumumab are mouse IgG2a. Second approved antibody-drug Second approved conjugate Other/comments Other/comments ascular thrombosis acute lymphoblastic acute leukemia anaplastic large cell lymphoma Cardiov Rheumatoid arthritis B-cell approved Obintuzumab is the first Autoimmune Alemtuzumab (Lemtrada) Multiple sclerosis Chronic lymphocytic Basiliximab ( Simulect)Daclizumab (Zenapax) (Blincyto) Multiple sclerosis rejection Transplant B-cell precursor Daclizumab: humanised IgG1k. (ReoPro) anti- Perisurgical (Gazyva) Ibritumomab tiuxetan (Zevalin) (Bexxar)* Tositumumab (Arzerra) Rituximab (Rituxan) (Adcetris)Brentuximab vedotin Hodgkin lymphoma, (Darzalex) (Darzalex) Daratumumab Multiple myeloma Bi-specific mAbs B B-lymphocyte subsets B-lymphocyte Alpha-chain of the IL-2 receptor killing of CD19 aggregation Platelet inhibitor differentiation receptor family leads to generation of leads to generation adenosine in the tumour microenvironment (immunosuppression); co-stimulatory receptor; more? Table 1. Targets and approvals for monoclonal antibodies and derivatives (asterisks denote approvals in USA only) only) in USA approvals denote (asterisks and derivatives monoclonal antibodies for and approvals Targets 1. Table CD52 of T- and Marker CD25 CD19/CD3 T-cell directed Bi-specific CD41 (glycoprotein llb/IIIa) CD20 in B-cell Participates CD30 Member of the TNF CD38 ADP ribose synthetase Cyclic Target Target Function mAbs, Traps, FDA-approved

222 © Royal College of Physicians 2017. All rights reserved.

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 222222 222/05/172/05/17 9:479:47 PMPM Monoclonal antibodies These mAbs designed not to be (eg no ADCC): IgG4. immune active This allows the mAbs to increase activity of T-cells without inducing for PDL-1 is a biomarker : IgG1k mAb engineered Atezolizumab: to lack ADCC activity intact (: PDL-1 is biomarker IgG1k)* tumour cells. IgG1 targets *Active likely Approval This is the first of the immuno-oncology This is the first (IgG1k). Largely mAbs to be approved of J Allison (ref PMID due to efforts 16730267) severe pain managed by morphine pain managed by severe Two monoclonal antibodies (T,P) and Two one ADC (ado-trastuzumab) of HER2 leads to Overexpression signaling spontaneous Crosslinking with mAb leads to internalization ADC First commercially successful PMID 19010901) et al, (Lewis therapy driven Biomarker Other/comments Other/comments ascular , non-small cell lung cancer, renal cell carcinoma, Hodgkin lymphoma, head and neck squamous carcinoma Urothelial (bladder) non-small carcinoma, merkel cell lung cancer cell carcinoma mesothelioma Breast, gastric adenocarcinoma Malignancy Cardiov Rheumatoid arthritis rejection Transplant Autoimmune Nolojix Nolojix fusion (both CTLA4-Fc Orencia proteins) (Opdivo) (Opdivo) Nivolumab (Keytruda) Pembrolizumab Atezolizumab (Tecentriq) Atezolizumab (Bavencio)* Avelumab Ipilimumb (Yervoy) melanoma, Metastatic (Unituxin) Neuroblastoma Mouse/human chimeric IgG1k. Induces Trastuzumab (Herceptin) Trastuzumab (Perjeta) emtansine Ado-trastuzumab (Kadcyla) Bi-specific mAbs B rovide co-stimulatory signals necessary for T-cell activation necessary for Ligand trap and survival. of CD28 activation prevents immune checkpoint resulting in immune suppression receptor that suppresses immune response after binding to PDL-1 or 2 that activates the PD-1 that activates immune suppression pathway binding CD80/86 this receptor turns down cytotoxic T-cells preferentially expressed preferentially on tumours of neuroectodermal origin without a known ligand (Continued) Table 1. Targets and approvals for monoclonal antibodies and derivatives (asterisks denote approvals in USA only) only) in USA approvals denote (asterisks and derivatives monoclonal antibodies for and approvals Targets 1. Table CD80/CD86 P CD279/PD-1 Immune ‘checkpoint’ CD274/PDL-1 Tumour-expressed protein CTLA4/CD152 through When activated Glycolipid Glycolipid GD2 Disialoganglioside HER2 receptor A growth factor-like Target Target Function mAbs, Traps, FDA-approved

© Royal College of Physicians 2017. All rights reserved. 223

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 223223 222/05/172/05/17 9:479:47 PMPM H Michael Shepard, Gail Lewis Phillips, Christopher D Thanos et al , are IgG1 ramucirumab Bevacizumab, (ramucirumab therapy cancer mAbs for phage display); from was derived (single fragment is a Fab arm binder). It has a short half-life but it intravenously, if administered when locally and effective is stable into the eye injected with is a ligand trap (Zaltrap) and cancer (Eylea) optical applications Biomarker: usually requires EGFR IHC Biomarker: for positivity; always requires test oncogene normal ras is mouse/human chimeric is human IgG2 from IgG1; Vectibix recombinant mice Not all TNF blockers are approved for all for are approved Not all TNF blockers indications are biosimilars: Drugs in italics adalimumab, infliximab, , inflectra adalimumab-atto are is a ; certolizumab pegol IgG1k mAbs is etanercept fragment; Fab pegylated that TNF receptor ‘trap’ a soluble Fc: binds TNF (there is one biosimilar) biosimilar TNF-blockers At least four in the EU (two been approved have infliximab and etanercept) each for Other/comments Other/comments ascular Colorectal cancer, cancer, Colorectal Non-squamous breast cancer, NSCLC, glioblastoma, renal gastric cell carcinoma, or gastro- cancer esophageal junction adenocarcinoma Squamous cell of the carcinoma head and neck, non- cancer, colorectal small cell lung cancer Malignancy Cardiov Age-related macular Age-related degeneration, macular oedema, diabetic macular oedema, Crohn's disease, colitis, RA, ulcerative idiopathicjuvenile arthritis, psoriatic arthritis, ankylosing spondylitis, plaque psoriasis, hidradentis uveitis suppurtiva, Autoimmune Bevacizumab (Avastin) Bevacizumab (Cyramza) Ramucirumab Aflibercept (Eylea/Zaltrap) (Lucentis) Ranibizumab Cetuximab (Erbitux) Cetuximab (Vectibix) Adalimumab (Humira) pegol (Cimzia) Certolizumab Golimumab (Simponi) Infliximab (Remicade) (Enbrel) Etanercept There are more than 20 anti- TNF biosimilars in various Already of development. stages are infliximab approved Inflectra, biosimilars (Remsina, biosimilars etanercept Flexiabi), (Erelzi, Benepali), adalimumab This field biosimilars (Amjevita). as many rapidly will change very dossiers are now under regulatory a scrutiny. Biosimilars are given (Erlezi) eg etanercept-szzs suffix, Bi-specific mAbs B vasculogenesis vasculogenesis and angiogenesis. in some Overproduced inflammatory disorders and tumours to induce increased blood supply. Many tumours are addicted Many tumours are addicted to the EGFR signalling pathway drives multiple autoimmune drives diseases* (Continued) Table 1. Targets and approvals for monoclonal antibodies and derivatives (asterisks denote approvals in USA only) only) in USA approvals denote (asterisks and derivatives monoclonal antibodies for and approvals Targets 1. Table VEGF that stimulates Cytokine EGFR (receptor multiple for growth factors) TNF-alpha Inflammatory cytokine that Target Target Function mAbs, Traps, FDA-approved

224 © Royal College of Physicians 2017. All rights reserved.

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 224224 222/05/172/05/17 9:479:47 PMPM Monoclonal antibodies κ IgE-related conditions (allergic rhinitis, IgE-related drug allergies, other) IgG4 natalizumab therapy has therapy IgG4 natalizumab with PML caused been associated John Cunningham in by immunocompromised patients. IgG1k not be associated MAY vedolizumab with PML. IgG2/4 mAb drug in the world Most expensive ($409,500 annually) Canakinumab is an IgG1k mAb; Canakinumab designed rilonacept is an IL-1 ‘trap’ with human mAb the IL-1R fused from region. Fc IgG1k human mAb from recombinant IgG1k human mAb from mice Human IgG2 mAb inhibits bone resorption and enhances bone mass (due to multiple causes) Murine/human chimeric IgG1 Other/comments Other/comments ascular ents destruction of red blood cells activated by complement (paroxysmal nocturnal hemoglobinuria) prevent bone mass prevent patients loss in cancer Castleman's Castleman's disease (similar to lymphoma) Malignancy Cardiov Multiple sclerosis, Crohn's disease and colitis ulcerative Rare inflammatory Rare active syndromes, arthritis, juvenile gouty arthritis arthritis, Crohn's disease Autoimmune Vedolizumab (Entyvio) Vedolizumab (Tysabri) Natalizumab Omalizumab (Xolair) Asthma IgG1k mAb also used off-label to treat Eculizumab (Solaris)Eculizumab Prev Canakinumab (Ilaris) Canakinumab Rilonacept (Arcalyst)* Ustekinumab (Stelara)Ustekinumab Plaque and psoriatic (Xgeva) be used to Can Siltuximab (Sylvant) Pseudo-malignancy: Bi-specific mAbs B Alpha-4 facilitates facilitates Alpha-4 integrin exit of inflammatory cells blood into intestine from or across the blood brain barrier basophils, other cells that receptor, express Fc-epsilon and induces release of inflammatory cytokines Inhibits complement cascade to overproduction of IL-1 to overproduction and inflammatory disease; other IL-1 also drives inflammatory diseases Prevents cytokine Prevents (inflammatory) activation of cellular receptors Activates osteoclastic bone osteoclastic Activates resorption associated with multiple associated malignancies (Continued) Table 1. Targets and approvals for monoclonal antibodies and derivatives (asterisks denote approvals in USA only) only) in USA approvals denote (asterisks and derivatives monoclonal antibodies for and approvals Targets 1. Table Alpha-4 integrin IgE Binds to mast cells, Complement Complement C5 -1 Interleukin-1 in cryopyrin lead Mutations P40 subunit of IL-12 and IL-23 Receptor Receptor of activator NF kappa-B ligand (RANKL) Interleukin-6 Interleukin-6 of IL-6 is Overexpression Target Target Function mAbs, Traps, FDA-approved

© Royal College of Physicians 2017. All rights reserved. 225

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 225225 222/05/172/05/17 9:479:47 PMPM H Michael Shepard, Gail Lewis Phillips, Christopher D Thanos et al Human/mouse chimeric mAb with vs RA after efficacy for initial approval of TNF blocker failure IgG1-gamma/lambda : IgG2 for low ADCC IgG2 for Evolocumab: is IgG1k Chimeric human/mouse chimeric mAb and prevention for to treatment SLAM7 receptor and to have a SLAM7 receptor and to have secondary mechanism of mediating cells ADCC vs multiple myeloma sequesters fragment This Fab blood, and clears it from dabigatran anti-coagulation reversing Other/comments Other/comments ascular Second line treatment for high cholesterol adults whose for is notcholesterol controlled by diet and statin treatment. Perhaps most important high hereditary for cholesterol Malignancy Cardiov polyarticular juvenile juvenile polyarticular arthritis, juvenile idiopathic arthritis erythematosus Autoimmune Tocilizumab (Actemra)Tocilizumab Rheumatoid arthritis, Belimumab (Benlysta) lupus Systemic (Anthim)* (Empliciti) Multiple myeloma This IgG1k mAb is thought to activate Evolocumab (Repatha) Evolocumab Alirocumab (Praluent) Bi-specific mAbs B Current approvals based Current approvals upon role of IL-6 in promoting inflammatory autoimmune disease Role in proliferation and in proliferation Role of B-cells differentiation Recognises the protective the protective Recognises antigen portion of the Bacillus toxin produced by the and prevents anthracis cells entering toxin from and killing them SLAM7 triggers and the activation of a wide differentiation of immune cells variety and adaptive (innate immune response) perhaps by primarily mediated killer cells and natural cells myeloma PCSK9 binds to LDLR, it to be degraded. causing With the result of higher of LDL mAb vs blood levels degradation PCSK9 prevents of LDLR and thereby from increases LDL removal blocking blood. Therefore, blood lower PCSK9 can levels cholesterol (Continued) Table 1. Targets and approvals for monoclonal antibodies and derivatives (asterisks denote approvals in USA only) only) in USA approvals denote (asterisks and derivatives monoclonal antibodies for and approvals Targets 1. Table IL-6R 6 (Interleukin receptor) BAFF (tumour necrosis factor superfamily member 13b) Bacillus anthraces (anthrax) toxin SLAMF7/ CD319 PCSK9 Dabigatran Dabigatran Anti-coagulant (Praxbind) Target Target Function mAbs, Traps, FDA-approved

226 © Royal College of Physicians 2017. All rights reserved.

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 226226 222/05/172/05/17 9:479:47 PMPM Monoclonal antibodies Ixekizumab is an IgG4; secukinumab an IgG1k Meant to be used for the prevention of the prevention Meant to be used for recurring clostridial disease. in the UK. is only approved of PDGF with its receptor(s) Other/comments Other/comments ascular ll activating factor; CTLA-4 = cytotoxic T-cell lymphocyte associated protein-4; EGFR = epidermal protein-4; associated CTLA-4 = cytotoxic T-cell lymphocyte factor; ll activating ty lipoprotein; LDR = low density lipoprotein receptor; NSCLC = non-small cell lung cancer; PCSK9 = non-small cell lung cancer; receptor; NSCLC LDR = low density lipoprotein ty lipoprotein; Malignancy Cardiov tural receptor for binding target; B bi-specific mAbs are engineered to bind to two different targets targets mAbs are engineered to bind two different bi-specific B binding target; receptor for tural lial growth factor lial growth factor PDL-1 = programmed cell death ligand 1; PML = progressive multifocal leukoencephalopathy; RA = leukoencephalopathy; multifocal cell death ligand 1; PML = progressive PDL-1 = programmed rug conjugate (X): toxin or radioisotope attached to mAb to increase efficacy. Asterisks denote approvals in approvals denote Asterisks to mAb increase efficacy. attached (X): toxin or radioisotope rug conjugate Asthma Both IgG1k mAbs Plaque psoriasis, ankylosing spondylitis Autoimmune Reslizumab (Cinqair) Reslizumab Mepolizumab (Nucala) Secukinumab (Cosentyx) (Zinplava) Actoxumab (Lartruvo)Olaratumab Soft tissue sarcoma interaction Human IgG1k that prevents Bi-specific mAbs B survival of eosinophils survival Clostridium difficile recurrence PDGF with its receptor (Continued) Table 1. Targets and approvals for monoclonal antibodies and derivatives (asterisks denote approvals in USA only) only) in USA approvals denote (asterisks and derivatives monoclonal antibodies for and approvals Targets 1. Table at www.antibodysociety.org. be found can summary of MAb approvals updated An actively with na receptors and compete from are derived traps T region), humanised or human; be murine, chimeric (human Fc MMabs can simultaneously (usually to bring immune cell into contact with target cell, thereby triggering target cell killing). Antibody d triggering target cell, thereby with target (usually to bring immune cell into contact simultaneously in both USA and UK. USA only; others are approved BAFF = B-ce cytotoxicity; ADP = adenosine diphosphate; ADCC = antibody-dependent cell-mediated ADC = antibody-drug conjugate; LDL = low densi receptor; HER2 = human EGFR-2; IHC immunohistochemistry; Ig immunoglobulin; IL interleukin; growth factor 1; cell death protein PD-1 = programmed growth factor; type 9; PDGF = platelet-derived subtilisin/kexin convertase = proprotein endothe VEGF = vascular molecule; TNF = tumour necrosis factor; activation rheumatoid arthritis; SLAMF7 = signalling lymphocyte IL-5 and Induces differentiation IL-17A Inflammatory cytokine Ixekizumab (Taltz) Clostridium recurrence of Reduce PDGF Inhibit association of the Target Target Function mAbs, Traps, FDA-approved

© Royal College of Physicians 2017. All rights reserved. 227

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 227227 222/05/172/05/17 9:479:47 PMPM H Michael Shepard, Gail Lewis Phillips, Christopher D Thanos et al

mAb trastuzumab (Herceptin®).20 The rigorous science that The strategy employed in the development of trastuzumab laid the foundation for this breakthrough mAb also initiated is now widely employed in cancer drug discovery (Fig 2 ). Very personalised/biomarker driven drug discovery and treatment recently in clinical trials, a combination of trastuzumab and in oncology. another anti-HER2 mAb (pertuzumab30 ) generated a complete Trastuzumab, the first successful monoclonal anti-cancer pathological response in approximately 50% of HER2-positive antibody to be successful against solid tumours, is well tolerated in patients,39 which is a previously unheard of therapeutic advance patients. The pathway leading to TNF-resistance of most tumour in the treatment of breast cancer. The HER2 programme cell lines 21 was unraveled by collaboration between the Shepard proved that mAbs have a place in therapy of solid tumours and (Genentech) and Schreiber laboratories (Chicago), which revealed that tyrosine kinase oncogenes were viable targets in cancer. that kill tumour cells largely by secreting TNF.22 Not all successful mAb therapeutics followed this pathway They hypothesised that if tumour resistance to macrophages of discovery. The first approved mAbs targeted membrane could be reversed, the tumour would become sensitive to killing proteins shared between haematologic malignancies and their by host defence . (or TNF)-resistant tumour cells precursor immune cell subsets (eg alemtuzumab (Campath®) implanted into syngeneic mice formed aggressive tumours, while – an anti-CD52, rituximab (Rituxan®) – an anti-CD20). These their TNF-sensitive parental cells regressed.21 mAbs ablate both normal and cancer cells. Alemtuzumab was The mechanism of TNF-resistance of tumours needed to the first humanised mAb therapeutic, depleting lymphocytes, be widespread since most tumour cell lines were resistant. monocytes and dendritic cells, useful in diseases such as Sporn and Todaro's autocrine growth factor hypothesis23 o f chronic lymphocytic leukemia. Similarly, rituximab depletes malignant transformation involving autocrine stimulation by CD20-expressing B-cells. Resistance to these mAb therapeutics transforming growth factors seemed plausible. Various growth occurs with the loss of the target as neither CD52 nor CD20 is factors were combined with TNF on TNF-sensitive tumour essential for malignant growth. These mAbs have toxicity issues cell lines and growth factors that activated receptor tyrosine due to massive depletion of immune cells, potentially causing kinases converted TNF-sensitive tumour cells to TNF-resistant ‘cytokine release syndrome’ or ‘tumour lysis syndrome’ – a cells.24 Host defence was completely subverted and the growth ‘cytokine storm’ resulting from aberrant immune activation inhibitor (TNF) even became a growth factor. 25 attributable to cellular debris activating ‘danger receptors’. Slamon et al 26,27 and Zhou et al28 reported that overexpression Despite these issues, both rituximab and alemtuzumab are of the HER2 in breast and ovarian successful drugs and often lead to disease regression. cancers was associated with more aggressive disease More recently, Jim Allison and other cancer researchers contemporaneously with Shepard's demonstration that discovered the utility of mAbs targeting ‘immune overexpression of HER2 induced resistance to TNF29 and that checkpoints’. By inhibiting these pathways (eg programmed mAbs to HER2 inhibited growth of HER2-overexpressing cell death protein 1 (PD-1)/programmed cell death ligand 1 tumours in vitro and in vivo .30–32 The lead antibody, MuMAb4D5 (PD-L1) or cytotoxic T-cell lymphocyte associated protein-4), – the murine partent of trastuzumab,24 w a s i n e f f e c t i v e o n tumour-associated immune suppression can be reversed with normal cells or tumour cells not overexpressing HER2.26,33 clinically effective immune cell attack on tumours in about Anti-HER2 also validated the importance of stability of the 20% cases. Some patients have had dramatic improvements, target itself as HER2 is indispensable in driving disease in notably in metastatic melanoma, and there is no doubt that subsets of breast, gastric and ovarian cancers. These findings this is a major step forward. Not unexpectedly there are laid the foundation for the success of trastuzumab and immune-mediated diseases in a large fraction (about 30%) subsequently for pertuzumab,34 which binds a separate epitope of treated patients, but this field of mAb therapy is growing on HER2, and then , an antibody drug very rapidly. CTLA-4 expressed on regulatory T-cells exerts conjugate. 35 The clinical success of trastuzumab required the immune suppressive activity. Anti-CTLA-4 prevents CTLA-4 co-development of two novel companion diagnostic approaches: from inhibiting immunity. 40 PD-1 is present on ‘exhausted’ immunohistochemistry and fluorescence in situ hybridisation for T-cells; anti-PD-1 reactivates them. The first approvals of selecting patients most likely to respond.36 This was a new idea at this new class of anti-cancer mAbs were in melanoma with the time, but is now a routine part of most cancer drug discovery (anti-CTLA-4, Yervoy®) and pembrolizumab and therapeutic programmes. Finally, this mAb showed that (anti-PD1, Keytruda®) in 2015, followed by atezolizumab antibody therapy was feasible in solid tumours, overcoming (anti-PD-L1, Tecentriq®) for non-small cell lung cancer and problems such as high internal tumour pressure, which causes bladder cancer. Checkpoint inhibitor mAbs, such as anti- vascular collapse and reverse flow of fluid in solid tumours.37 OX40, can also be agonistic.40 MuMAb4D5 was humanised using a consensus human variable domain sequence from the Kabat database, together Other antibody-related approaches and mechanisms with human immunoglobulin constant domain (IgG1, kappa) sequences. 38 This resulted in the synthesis of a recombinant Therapeutic antibodies: structural marvels that mediate antibody, non-existent in nature, with almost 95% human multiple functional effects sequence, reducing concerns related to immunogenicity. The Antibodies have evolved over the past 400 million years to humanised antibody has three mechanisms of action dependent provide broad ranging host defence in all vertebrates. Thus, it upon upregulated HER2: is not surprising that engineered antibodies have emerged as 1 growth inhibition one of the main sources of new medicines. Antibodies evolved 2 antibody-dependent cell-mediated cytotoxicity (ADCC) to possess several important functional properties exploited 3 induction of TNF sensitivity. 20,30 by recombinant mAb therapeutics, such as exquisite specificity

228 © Royal College of Physicians 2017. All rights reserved.

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 228228 222/05/172/05/17 9:479:47 PMPM Monoclonal antibodies

Goal: Discover a target for cancer therapy that can inhibit tumour progression, but not injure normal ssues

Proof of concept in disease models in vivo: Target discovery strategy: Ulise tumour cell lines that overexpress p185-HER2 to Because macrophages preferenally kill tumour cells and create xenogra s and show that monoclonal anbody was most tumour cells are resistant to macrophage/TNF- efficacious at doses translatable to clinical use. Unlike many mediated killing, base target discovery on the underlying tumour markers, p185-HER2 is stable and loss of receptor mechanisms of resistance to TNF-macrophage killing expression by a tumour leads to loss of tumourigenicity

Demonstrate therapeuc modality in vitro: Key pathway discovery: Show that monoclonal anbody versus overexpressed Discovery that growth factor-mediated tyrosine kinase p185-HER2 could inhibit tumour cell growth and induce acvaon in macrophage/TNF-sensive cells induced a sensivity to macrophage/TNF toxicity in vitro macrophage/TNF-resistant phenotype

In vitro target proof of concept: Demonstrate that overexpressed p185-HER2 results in resistance to macrophage/TNF killing and may provide mechanisms underlying more aggressive cancer in tumours that overexpress p185-HER2

Fig 2. Strategy and key steps for development of trastuzumab, from target discovery to in vitro and in vivo proof of concept studies. HER2 = human receptor 2; TNF = tumour necrosis factor

and affinity toward their antigen targets, recruiting immune and then ‘humanised’ where all but the binding site to the HER2 system effector components and a long serum half-life. antigen was changed to a human sequence (Fig 1 ). Genetically Antibodies possess a unique tertiary domain structure, engineered mice have been developed that can make fully human folding into a complex quaternary structure. Human serum antibodies, eg Vectibix® (panitumumab) – anti-epidermal immunoglobulin is approximately 80% IgG, as are most growth factor receptor. Another important approach employs approved therapeutic antibodies. These complex proteins synthetic (human) antibody libraries that are displayed on the (approximately 150 kDa) comprise four polypeptide chains – 2 surface of yeast or phage, which is useful for less immunogenic heavy and 2 light chains – with disulphide bonds providing targets. There are advantages and limitations to each of these significant rigidity. The heavy chain consists of a variable methods; therefore, antibody discovery groups will attempt domain (VH) and three constant domains (CH1, CH2 and multiple approaches in parallel against a particular target. CH3). The light chain consists of a variable domain (VL) and a constant domain (CL) (Fig 1 A). IgG antibodies are bivalent and Antibody effector function have post-translational modifications. Glycosylation in the Fc Immunoglobulins can stimulate immune defence mechanisms domain also helps to provide stability and modulates properties 41 through ADCC, antibody-dependent cellular phagocytosis such as binding to Fc receptors. and complement-dependent cytotoxicity. During ADCC, immune cells – usually natural killer (NK) cells – lyse a The antibody/antigen interaction target cell with an IgG bound to a cell surface target. CD16 Fc Several technologies are used to identify novel antibodies: receptors expressed on NK cells bind to the Fc region of the hybridomas, genetically modified mice with human IgG, forming a lytic synapse between the NK cell and the target immunoglobulin sequences and phage or yeast display. cell, destroying the target cell. During complement-dependent B-cells from a mouse injected with an antigen are fused with cytotoxicity, complement is recruited to the IgG-bound surface immortalised B-cell myeloma cells to make hybridomas. To pathogen through C1q binding to the antibody Fc domain, improve their efficacy in patients, protein engineers created triggering proteolytic events, yielding a membrane attack chimeric antibodies containing human constant domains and complex that disrupts cellular membranes with cell death. murine variable domains, which reduce immunogenicity and Many therapeutic antibodies use ADCC as a key mechanism activate effector functions (Fig 1 ). Approved chimeric antibodies of action, eg trastuzumab and rituximab. Methods have been include Remicade® (infliximab), Erbitux® (cetuximab) and developed for increasing or decreasing antibody effector Rituxan® (rituximab). As an example of a ‘humanised’ antibody function, the number of epitope binding sites or attaching Herceptin® (trastuzumab) was isolated from a murine hybridoma cytotoxic drugs. Some of these strategies are shown in Fig 1 .

© Royal College of Physicians 2017. All rights reserved. 229

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 229229 222/05/172/05/17 9:479:47 PMPM H Michael Shepard, Gail Lewis Phillips, Christopher D Thanos et al

AB C

Phage CD19 angen-binding region plus linker aached to CD3 angen-binding region scFv VH VH

VL VL scFv Linker Camelid anbodies

Bi-specific T-cell engager (BiTE)

Fab IgG CD3 BiTE Tumour angen eg CD19

F-Cab

Fig 3. Emerging antibody technologies. A – derivatives of classical mAbs: scFv, Fab, and F-Cab antibody formats; B – engineered bi-specifi c mAbs: structure and mechanism of a bi-specifi c T-cell engaging (BiTE) antibody; C – novel mAb frameworks: unique structure of camelid antibodies. mAb = monoclonal antibody

Antibody Despite decades of intense research and development, Antibodies have a long serum half-life, typically lasting only three ADCs have gained marketing approval. The first, 2–4 weeks in circulation. Antibody binding the Fc domain of (Mylotarg®), was approved then the neonatal , FcRn, recycles the antibody, protecting taken off the market. Significant advances were subsequently it from other clearance mechanisms. This long half-life with made in ADC linker design and conjugation technologies, less frequent dosing is often more favourable for patients when yielding two additional approved ADCs, one for CD30- extended drug function is needed. Antibody dosing is typically positive Hodgkin lymphoma and anaplastic large cell intravenous or subcutaneous. Oral administration is not lymphoma (brentuximab vedotin (Adcentris®), and one feasible because of rapid degradation of the antibody in the gut. for metastatic breast tumours overexpressing HER2/neu In addition, therapeutic antibodies cannot pass in appreciable trastuzumab emtansine (Kadcyla®). The anti-CD30 antibody quantities through the intact blood-brain barrier. component of brentuximab vedotin is linked to the tubulin polymerisation inhibitor mono-methyl auristatin E through Antibody-drug conjugates: using antibodies for tumour- a protease cleavable linker, valine-citrulline. Trastuzumab selective delivery of cytotoxics emtansine is comprised of trastuzumab covalently linked to the maytansinoid derivative, DM1, through a stable, non-cleavable The therapeutic use of antibody-drug conjugates (ADCs) is linker. Current research suggests that more efficacious and based on antibody-mediated tumour-selective delivery of better tolerated ADCs will be developed, which will provide potent cytotoxic compounds. This relies on antibody-induced greater clinical benefit to cancer patients. receptor internalisation, followed by trafficking of the ADC to the lysosomes where the cytotoxic drug is released from the Emerging antibody technologies ADC. The free drug (inside the target cell) initiates its anti- tumour activity. Stability of the linker that joins the antibody The Fab fragment (Fig 3 A) used in ranibizumab (Lucentis) 42 and cytotoxic agent is key to both efficacy and safety. The can be manufactured in Escherichia coli. The short half- linker must be stable in the circulation to prevent premature life of Fabs can be remedied by polyethylene glycol used in release of the cytotoxic agent, yet allow release of the cytotoxic certilizumab pegol, Cimzia®.43 drug within the tumour cell. The most common linkers are Until recently, it has been difficult to synthesise bi-specific cleavable linkers such as peptides, cleaved by proteases, or mAbs. Now various types are possible. Bi-specific T-cell disulphide linkers, which undergo reduction to release the engaging (BiTE) antibodies consist only of the variable regions drug within the lysosome. ADCs can also utilise a stable or from distinct antibodies (Fig 3 B) and are the format of the first non-cleavable linker, the free drug is released in the lysosome approved bi-specific antibody, (Removab®), for after lysosomal degradation of the entire ADC molecule. malignant ascites.

230 © Royal College of Physicians 2017. All rights reserved.

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 230230 222/05/172/05/17 9:479:47 PMPM Monoclonal antibodies

Antibody-based cellular immunotherapies 6 Choy EH , Kingsley GH , Panayi GS . Anti-CD4 monoclonal anti- bodies in rheumatoid arthritis . Springer Semin Immunopathol A new class of antibody-based therapeutics called ‘chimeric 1998 ; 20 : 261 – 73 . antigen receptor T-cells’ are emerging in cancer. An antibody- 7 Van Wauwe JP , De Mey JR , Goossens JG . OKT3: a monoclonal anti- like antigen binding region is fused with signalling components human T lymphocyte antibody with potent mitogenic properties. J of the T-cell receptor and the chimeric gene is inserted into the Immunol 1980 ; 124 : 2708 – 13 . patient's own T-cells. When the T-cells recognise their target on 8 Feldmann M , Maini RN . Lasker Clinical Medical Research Award. tumour cells, the T-cell cytotoxic response is activated, lysing TNF defined as a therapeutic target for rheumatoid arthritis and the cancer cells.43 Using CTL019, a chimeric antigen receptor other autoimmune diseases . Nature Medicine 2003 ; 9 : 1245 – 50 . 9 Brennan FM , Chantry D , Jackson A , Maini RN , Feldmann M . T-cell engineered to target cancer cells that express the CD19 Inhibitory effect of TNF antibodies on synovial cell interleukin-1 protein present on acute lymphoblastic leukemia cells, 19 of 22 production in rheumatoid arthritis. Lancet 1989 ; 2 : 244 – 7 . paediatric patients with lethal acute lymphoblastic leukaemia 10 Leombruno JP , Einarson TR , Keystone EC . The safety of anti- 44 experienced remission. However, this treatment upregulates tumour necrosis factor treatments in rheumatoid arthritis: meta inflammatory cytokines, with consequent toxicity. and exposure-adjusted pooled analyses of serious adverse events. Ann Rheum Dis 2009 ; 68 : 1136 – 45 . Conclusions 11 Kievit W , Fransen J , Adang EM et al . Long term effectiveness and safety of TNF-blocking agents in daily clinical practice: results from mAb therapeutics have been approved for 33 targets in more the Dutch Rheumatoid Arthritis Monitoring register. Rheumatol than 37 distinct diseases (Table 1), most commonly for 2011 ; 50 : 196 – 203 . cancer (27 approvals). The clinical value of both mAbs and 12 van der Bijl AE , Goekoop-Ruiterman YP , de Vries-Bouwstra ligand traps has been proven. The most targeted pathway J K, et al. Infliximab and methotrexate as induction therapy in patients with early rheumatoid arthritis. Arthr Rheum is the TNF pathway, now employed to treat nine different 2007 ; 56 : 2129 – 34 . indications and likely to expand as TNF is found to be a driver 13 Quinn MA , Conaghan PG , O'Connor PJ et al . Very early treat- of additional diseases and conditions, including postoperative ment with infliximab in addition to methotrexate in early, 45 46 cognitive decline and fibrosis. New applications of mAbs poor-prognosis rheumatoid arthritis reduces magnetic reso- 47 are being tested, with approvals in bone metabolism a n d nance imaging evidence of synovitis and damage, with sustained hypercholesterolemia.48 benefit after infliximab withdrawal. Results from a 12-month Antibodies have become the new backbone of the randomized, double-blind, placebo-controlled trial. Arthr Rheum pharmaceutical industry, which previously relied on huge 2005 ; 52 : 27 – 35 . libraries of small molecules. Compared with small molecules, 14 Kang S , Tanaka T , Kishimoto T . Therapeutic uses of anti-inter- mAbs have an exquisite target selectivity and, therefore, less leukin-6 receptor antibody . Int Immunol 2015 ; 27 : 21 – 9 . 15 Cohen MD , Keystone E . Rituximab for rheumatoid arthritis . toxicity attributable to binding to other targets. mAbs have now Rheumatol Ther 2015 ; 2 : 99 – 111 . been designed to target two or more targets simultaneously, 16 Hahn BH. Belimumab for systemic lupus erythematosus . New Engl augmenting the therapeutic potential. We are only just at the J Med 2013 ; 368 : 1528 – 35 . beginning of the mAb therapeutic era. ■ 17 Feagan BG , Sandborn WJ , Gasink C et al. Ustekinumab as induc- tion and maintenance therapy for Crohn's Disease. New Engl J Med C o n fl i c t s o f i n t e r e s t 2016 ; 375 : 1946 – 60 . 18 Baeten D , Sieper J , Braun J et al. Secukinumab, an interleukin-17A GLP is an employee of Genentech and owns Roche stock. CT and HMS inhibitor, in Ankylosing Spondylitis . 2015 ; New Engl J Med. 373 : are employees of Halozyme and own Halozyme stock. MF is one of the 2534 – 48 . inventors of anti-TNF therapy and receives royalties on combination 19 Weinblatt ME , Moreland LW , Westhovens R et al . Safety of abata- therapy. cept administered intravenously in treatment of rheumatoid arthritis: integrated analyses of up to 8 years of treatment from the Author contributions abatacept program. J Rheumatol 2013 ; 40 : 787 – 97 . 20 Carter P , Presta L , Gorman CM et al. Humanization of an anti- All authors wrote part of the review, read all of it and approved the p185HER2 antibody for human cancer therapy . Proc Natl Acad Sci fi n a l v e r s i o n . USA 1992 ; 89 : 4285 – 9 . 21 Sugarman BJM , Aggarwal BB , Hass PE et al. Recombinant human tumour necrosis factor-alpha: effects on proliferation of normal References and transformed cells in vitro . Science 1985 ; 230 : 943 – 5 . 1 Köhler G , Milstein C . Derivation of specific antibody-producing 22 Urban JL , Shepard HM , Rothstein JL , Sugarman BJ , Schreiber tissue culture and tumor lines by cell fusion. Eur J Immunol H . Tumor necrosis factor: a potent effect molecule for tumor 1976 ; 6 : 511 – 9 . cell killing by activated macrophages . Proc Natl Acad Sci USA 2 Morrison SL , Johnson MJ , Herzenberg LA , Oi VT . Chimeric human 1986 ; 83 : 5233 – 7 . antibody molecules: mouse antigen-binding domains with human 23 Sporn MB , Todaro GJ . Autocrine secretion and malignant transfor- constant region domains . Proc Natl Acad Sci USA 1984 : 81 : 6851 – 2 . mation of cells. N Engl J Med 1980 ; 303 : 878 – 80 . 3 Jones PT , Dear PH , Foote J , Neuberger MS , Winter G . Replacing 24 Sugarman BJ , Lewis GD , Eessalu TE , Aggarwal BB , Shepard HM . the complementarity-determining regions in a human antibody Effects of growth factors on the anti proliferative activity of tumor with those from a mouse. Nature 1986 ; 321 : 522 – 5 . necrosis factors . Cancer Res 1987 ; 47 : 780 – 6 . 4 An Z. Monoclonal antibodies – a proven and rapidly expanding 25 Lewis GD , Aggarwal BB , Eessalu TE , Sugarman BJ , Shepard therapeutic modality for human diseases. Protein Cell 2010 ; 1 : 319 – 30 . HM . Modulation of the growth of transformed cells by human 5 Burnet FM. Immunological recognition of self. Science tumor necrosis factor-alpha and interferon-gamma. Cancer Res 1961 ; 133 : 307 – 11 . 1987 ; 47 : 5382 – 5 .

© Royal College of Physicians 2017. All rights reserved. 231

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 231231 222/05/172/05/17 9:479:47 PMPM H Michael Shepard, Gail Lewis Phillips, Christopher D Thanos et al

26 Slamon DJ , Clark GM , Wong SG et al . Human breast cancer: corre- 39 Tiwari SR , Mishra P , Raska P et al . Retrospective study of the lation of relapse and survival with amplification of the HER-2/neu efficacy and safety of neoadjuvant docetaxel, carboplatin, trastu- oncogene. Science 1987 ; 235 : 177 – 82 . zumab/pertuzumab (TCH-P) in nonmetastatic HER2-positive 27 Slamon DJ , Godolphin W , Jones LA et al . Studies of the HER-2/ breast cancer . Breast Cancer Res Treat 2016 ; 258 : 189 – 93 . neu proto-oncogene in human breast and ovarian cancer . Science 40 Mahoney KM , Rennert PD , Freeman GJ . Combinatino cancer 1989 ; 244 : 707 – 12 . immunotherapy and new immunomodulatory targets. Nat Rev 28 Zhou D , Battifora H , Yokota J , Yamamoto T , Cline MJ . Association Drug Discov 2015 ; 14 : 561 – 84 . of multiple copies of the c-erbB-2 oncogene with spread of breast 41 Bournazos S , Ravetch JV . Fc receptor pathways during active and cancer. Cancer Res 1987 ; 47 : 6123 – 5 . passive immunization . Immunol Rev 2015 ; 268 : 88 – 103 . 29 Hudziak RM , Lewis GD , Shalaby MR et al . Amplified expres- 42 Ferrara N , Damico L , Shams N , Lowman H , Kim R . Development sion of the HER2/ERBB2 oncogene induces resistance to tumor of ranibizumab, an anti-vascular endothelial growth factor antigen necrosis factor alpha in HIH 3T3 cells . Proc Natl Acad Sci USA binding fragment, as therapy for neovascular age-related macular 1988 : 85 : 5102 – 6 . degeneration . Retina 2006 ; 26 : 859 – 70 . 3 0 L e w i s G D, F i g a r i I , F e n d l y B et al . Differential responses of human 43 Choy EH , Hazleman B , Smith M et al. Efficacy of a novel tumor cell lines to anti-p185HER2 monoclonal antibodies . Cancer PEGylated humanized anti-TNF fragment (CDP870) in patients Immunol Immunother 1993 ; 37 : 255 – 63 . with rheumatoid arthritis: a phase II double-blinded, randomized, 3 1 C e r a n C , C o k o l M , C i n g o z S et al. Novel anti-HER2 monoclonal dose-escalating trial. Rheumatology 2002 ; 41 : 1133 – 7 . antibodies: synergy and antagonism with tumor necrosis factor- 44 Maude SL , Frey N , Shaw PA , Aplenc R , Barrett DM . Chimeric alpha. BMC Cancer 2012 ; 12 : 450 . antigen receptor T cells for sustained remissions in leukemia. N 32 Sobol RE , Astarita RW , Hofeditz C et al. Epidermal growth factor Engl J Med 2014 ; 371 : 1507 – 17 . receptor expression in human lung carcinomas defined by a mono- 4 5 Te r r a n d o N , M o n a c o C , M a D et al. Tumor necrosis factor-alpha clonal antibody . J Natl Cancer Inst 1987 : 79 : 403 – 7 . triggers a cytokine cascade yielding postoperative cognitive decline. 33 Varley JM , Swallow JE , Brammer WJ , Whittaker JL , Walker RA . Proc Natl Acad Sci USA 2010 ; 107 : 20518 – 22 . Alternations to either c-erbB-2(neu) or c-myc proto-oncogenes 46 Glass GE , Chan JK , Freidin A et al. TNF promotes fracture repair in breast carcinomas correlate with poor short-term prognosis. by augmenting the recruitment and differentiation of muscle- Oncogene 1987 ; 1 : 423 – 30 . derived stromal cells. Proc Natl Acad Sci USA 2010 ; 108 : 1585 – 90 . 34 Adams CW , Allison DE , Flagella K et al . Humanization of a 47 Pageau SC. Denosumab. MAbs 2009 ; 1 : 210 – 5 . recombinant monoclonal antibody to produce a therapeutic HER 48 Everett BM , Smith RJ , Hiatt WR . Reducing LDL with PCSK9 dimerization inhibitor, pertuzumab. Cancer Immunol Immunother inhibitors - the clinical benefit of lipid drugs. N Engl J Med 2006 ; 55 ; 717 – 27 . 2015 ; 373 : 1588 – 91 . 35 Lewis Phillips GD , Li G , Dugger DL et al . Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res 2008 ; 68 : 9280 – 90 . 36 Lebeau A , Deimling D , Kaltz C et al . Her-2/neu analysis in archival tissue samples of human breast cancer: comparison of immuno- histochemistry and fluorescence in situ hybridization. J Clin Oncol Address for correspondence: Professor Sir Marc Feldman, 2001 ; 19 : 354 – 63 . Kennedy Institute of Rheumatology, Botnar Research 37 Jain RK. Vascular and interstitial barriers to delivery of therapeutic Centre, Nuffield Dept of Rheumatology Orthopedics and agents in tumors . Cancer Rev 1990 ; 9 ; 253 – 66 . Musculoskeletal Science, University of Oxford, Windmill Road, 38 Johnson G , Wu TT . Kabat database and its applications: 30 years Headington, Oxford OX3 7LD. after the first variability plot . Nucleic Acids Res 2000 ; 28 : 214 – 8 . Email: [email protected]

232 © Royal College of Physicians 2017. All rights reserved.

CCMJv17n3-Feldman.inddMJv17n3-Feldman.indd 232232 222/05/172/05/17 9:479:47 PMPM