Key Points
- •
Biologics are mostly large molecules, usually proteins, that serve as therapeutics and are derived from living organisms.
- •
Biologics produced in vitro by recombinant technology are monoclonal antibodies, soluble receptor constructs, or cytokines. The first two categories are discussed in this chapter.
- •
Recombinant biologics bind their target with high affinity, and have reduced potential for off-target effects compared to small molecule agents.
- •
Safety concerns with biologics arise from their effectiveness in targeting physiologic molecules or cells, or from their ability to elicit an immune response against the drug itself.
- •
The molecular complexity and unavoidable heterogeneity of recombinant biologics make them expensive to produce and impossible to copy exactly in a generic form. In the future, biosimilars will be produced that mimic an existing approved biologic as far as possible.
Biologics are drugs that are derived from living organisms. In the broadest sense, this category encompasses any agent made by biological processes, including recombinant therapeutic proteins, as well as blood, blood products, proteins or other molecules purified from living material, such as IVIG (see Chapter 15 ), cells and gene therapies ( Chapter 16 ), allergens ( Chapter 23 ) and vaccines. We discuss in this chapter therapeutic proteins that are monoclonal antibodies or soluble receptor constructs. They have all been developed by genetic engineering, and they are produced in vitro in large cultures of prokaryotic or eukaryotic cells. Therapeutic cytokines manufactured by recombinant technology comprise another increasingly important subset of biologics. They are listed in Table 17-1 but will not be further described here.
Compound | Brand Name, Manufacturer | Pediatric Indications | Age Restriction |
---|---|---|---|
C1 esterase inhibitor | Cinryze ® | Hereditary angioedema | Adolescents |
Epoetin alfa | Epogen ® | Anemia | None |
Filgrastim (G-CSF) | Neupogen ® | Neutropenia | None |
Interferon alfa-2b | Intron ® A | Chronic hepatitis B | ≥ 1 yr |
Interferon gamma | Actimmune ® | Chronic granulomatous disease, osteopetrosis | None |
Peginterferon alfa-2a | Pegasys ® | Chronic hepatitis C | ≥ 5 yr |
Peginterferon alfa-2b | Pegintron ® | Chronic hepatitis C | ≥ 3 yr |
Sargramostim | Leukine ® | Bone marrow transplantation, chemotherapy | None |
The fundamental therapeutic rationale underlying the biologics discussed in this chapter relates to the exquisite specificity of the humoral immune system’s antibodies or of certain physiologic receptor-ligand interactions, coupled with the identification of molecules or cells that are critical to the pathologic processes of disease. This permits the development of agents that will target selected steps in a disease pathway with very high affinity yet have little propensity to have ‘off-target’ interactions that could lead to serious side-effects. In addition to their specificity, many of the biologics closely resemble physiologic molecules, and as such, they may be administered in large quantities to achieve the substantial levels (hundreds of micrograms per mL) that can potentially block all of their targets in vivo. This pharmacodynamic efficiency of the biologics certainly accounts for their impressive efficacy in many uses. It also implies that the failure of a particular biologic to provide therapeutic benefit in a given situation makes it unlikely that the targeted molecule plays a non-redundant role in the disease process.
The complex nature of biologics in part accounts for their high cost, which limits their general use, particularly in developing countries. It also presents several other potential problems particular to this group of drugs. Their molecular complexity makes them relatively difficult to develop and often entails a degree of structural heterogeneity that may have unintended consequences. Any change in the manufacturing process must be assumed to change the final product to some extent. This makes the generation of generic forms of biologics essentially impossible. So-called ‘biosimilars’ can have the same amino acid sequence as the first-approved biologic, but the post-translational modifications will not be identical. This variability demands that the regulatory requirements for approval must go beyond any demonstration of chemical identity and should include some biological measures of equivalence in pharmacokinetics, pharmacodynamics, clinical efficacy and safety. Such requirements would need to be less stringent than those applied to a new compound, or the potential financial benefit of the biosimilar (i.e. lower patient cost) would be abrogated. Biosimilar mimics of the compounds discussed in detail in this chapter have not yet appeared in the US market, and the path to approval will have to be developed individually in each case. Compounds that have the same targets but are independently developed (e.g. adalimumab and infliximab), are of course not biosimilars, and each has to undergo its own extensive developmental process. Nevertheless, the success and failures of a given compound will help guide the clinical development of others with the same target. In addition, the safety concerns that the US Food and Drug Administration (FDA) requires in the package inserts may reflect analogies with other compounds with the same target, even in the absence of adverse event data that are directly relevant.
Another general concern about biologics is immunogenicity. As large protein molecules, these drugs are potentially good immunogens. Most of the receptor constructs, such as rilonacept, are different from any autologous protein in the overall structure (anakinra is the exception), although they are made up of faithful portions of individual physiologic molecules. The monoclonal antibodies are more physiologic, but they also can provoke immune responses. Muromonab-CD3, the first monoclonal antibody approved in the USA in 1986, was derived from a mouse immunized with human T cells. It recognizes the CD3ε subunit of the T cell receptor-CD3 complex, and it is effective in treating allograft rejection. However, it frequently induces anti-mouse IgG antibodies (termed HAMA, or human anti-mouse antibodies), which is part of the reason it was withdrawn from the market in 2010. Some murine monoclonal antibodies are still used clinically (e.g. ibritumomab in adults) for short courses of therapy, but in general, further genetic modifications are undertaken to render the monoclonal more human and thus, less immunogenic.
A monoclonal antibody is usually selected from a mouse immunized with the appropriate human cells or protein. The variable regions of both the light and heavy chains are then joined, respectively, to the constant region domains of human κ or λ, and IgG (IgG1, IgG2 or IgG4) to a make a chimeric molecule that is about two thirds of human origin. Infliximab is an example. Such proteins can still not infrequently induce antibody responses, termed HACA, or human anti-chimera antibodies. Further humanization can be obtained by substituting the framework regions in the variable domains with human framework sequences. This leaves only the complementarity determining regions (hypervariable regions) from the original mouse monoclonal. These are the sequences that determine the antigen-binding site, so the specificity of the engineered monoclonal remains the same, but it now has only about 10% mouse sequences. An example of such a humanized monoclonal is eculizumab.
A fully human monoclonal antibody can be produced in two ways. In one approach, a mouse is used in which the immunoglobulin genes have been replaced with human genes. This mouse then makes antibody responses that use human antibody sequences. Canakinumab was produced in this way. A second approach uses a bacteriophage display library of human variable region sequences and selects for antigen binding in vitro. The selected genes are then combined with human constant region genes to reconstitute a complete IgG antibody. Adalimumab was derived in this manner. Notwithstanding, these human monoclonals can provoke immune responses (termed HAHA, or human anti-human antibody), probably largely akin to anti-idiotype responses that are themselves specific for the reagents’ complementarity determining regions. Table 17-2 describes how the mouse/human composition of the monoclonal antibody produced is reflected in the spelling of the penultimate and antepenultimate syllables of its common name.
|
* Reproduced from: Stiehm’s Immune Deficiencies , Edited by KE Sullivan and ER Stiehm, Elsevier, Amsterdam, 2014, page 891.
The immune responses to biologic agents that are often seen in a significant minority of patients may hasten the clearance of the drug, decrease its effectiveness or result in adverse reactions such as hypersensitivity. In many cases, however, they do not seem to have apparent clinical significance, and their presence is not assayed in routine clinical care. In the early studies with infliximab, it was found that higher doses of drug, or giving it along with methotrexate, decreased the incidence of HACA formation. It is not known how well this paradigm may extend to other compounds.
Another class of adverse reactions to the biologics comes from their therapeutic potency. Although they are unlikely to have ‘off-target’ side-effects, they are often so thorough in inhibiting their target that the normal physiologic functions of that target are blocked. Thus, the anti-TNF agents can reactivate latent mycobacterial infections in a characteristic way, or eculizumab (anti-C5) can put patients at risk for serious infections with Gram-negative cocci by blocking the terminal complement cascade.
The compounds discussed below are biologic monoclonal antibodies or receptor constructs that have an FDA-indicated use in the pediatric population as of mid-2014 (see Table 17-3 ). Other biologics approved only for adults have found common use in children. Some of them are listed in Table 17-4 , but they will not be further discussed here.
Compound | Target | Pediatric Indications | Age Restrictions | FDA Approval ‡ |
---|---|---|---|---|
Abatacept | CD80, CD86 | Juvenile idiopathic arthritis | ≥ 6 yr | 2008 |
Adalimumab | TNF-α | Juvenile idiopathic arthritis | ≥ 4 yr | 2008 |
Anakinra | IL-1R | Neonatal onset multisystem inflammatory disease | None | 2014 |
Basiliximab | CD25 | Renal transplantation | None | 2001 |
Canakinumab | IL-1β | Familial cold autoinflammatory syndrome , Muckle-Wells syndrome , SJIA | CAPS ≥ 4 yr; SJIA ≥ 2 yr | 2009 |
Denosumab | RANKL | Giant cell tumor | Bone-mature adolescents | 2013 |
Ecallantide | Kallikrein | Hereditary angioedema | ≥ 12 yr | 2009 |
Eculizumab | C5 | Atypical hemolytic uremic syndrome | None | 2009 |
Etanercept | TNF-α, TNF-β | Juvenile idiopathic arthritis | > 2 yr | 1999 |
Infliximab | TNF-α | Inflammatory bowel disease | ≥ 6 yr | 2006 |
Omalizumab | IgE | Asthma, chronic idiopathic urticaria | ≥ 12 yr | 2003 |
Palivizumab | RSV F protein | RSV prevention | ≤ 24 months | 1998 |
Raxibacumab | Anthrax toxin | Anthrax | None | 2012 |
Rilonacept | IL-1β | Familial cold autoinflammatory syndrome , Muckle-Wells syndrome | ≥ 12 yr | 2008 |
Tocilizumab | IL-6R | Polyarticular and systemic juvenile idiopathic arthritis | ≥ 2 yr | 2011 |
* These compounds are discussed in more detail in the text.
‡ Year that FDA approval was first granted for a pediatric indication.
Compound | Brand Name | Target | Pediatric Uses |
---|---|---|---|
Abciximab | ReoPro ® | GpIIb/IIIa receptor | Kawasaki disease |
Alemtuzumab | Campath ® | CD52 | Hemophagocytic lymphohistiocytosis |
Belimumab | Benlysta ® | BLyS | SLE |
Bevacizumab | Avastin ® | VEGF-A | Retinopathy of prematurity, malignancies |
Golimumab | Simponi ® | TNF-α | Juvenile idiopathic arthritis, uveitis |
Natalizumab | Tysabri ® | α4 integrin subunit | Multiple sclerosis |
Rituximab | Rituxan ® | CD20 | SLE, transplant, lymphoma/leukemia, others |
Ustekinumab | Stelara ® | IL-12, IL-23 | Psoriasis |
* These compounds are not discussed in detail in the text. They are all the subject of ongoing (2014) clinical trials in children for the indicated uses, except abciximab, which is listed as being studied only for sickle cell crisis in children ( http://www.clinicaltrials.gov ).
Biologics Approved for Pediatric Therapeutic Use
Abatacept (Orencia ® ).
Abatacept is a soluble receptor mimic that consists of the extracellular domain of CTLA-4 (cytotoxic T lymphocyte antigen-4) linked to the Fc fragment of human IgG1. It is produced in vitro from a chimeric gene construct. CTLA-4 is found on activated CD4 and CD8 T cells, and it mediates an inhibitory signal through ligation to CD80 or CD86. However, the mechanism of action of abatacept results from its high-affinity binding to CD80 and CD86, which thereby prevents their interaction with the costimulatory molecule CD28, which is in turn constitutively expressed on CD4 and CD8 T cells. It was initially approved for use in adult rheumatoid arthritis in 2005. The pediatric indication for children 6 to 17 years old with juvenile idiopathic arthritis was added in 2008 on the basis of a 6-month withdrawal trial using 122 patients with polyarticular disease. These patients were also found to benefit in a longer-term open-label follow-up. Abatacept is administered intravenously once a month, either as monotherapy or in conjunction with methotrexate. A preparation for subcutaneous self-administration is available only for adult use. Infections are a potential complication with abatacept therapy, although in general the risk may be less than with other biologics used for inflammatory arthritis. Hepatitis B reactivation, however, may be a particular concern.
Adalimumab (Humira ® ).
Adalimumab is a fully human recombinant monoclonal antibody that binds the soluble and cell-bound forms of TNF-α. It was initially approved for adult rheumatoid arthritis in 2002. An indication for juvenile idiopathic arthritis in children at least 4 years old was added in 2008 on the basis of a 32-week withdrawal study of 128 patients with polyarticular disease. It is administered in weekly or biweekly subcutaneous doses, either as monotherapy or in combination with methotrexate. In adults (18 years and older), it is also approved for use in ankylosing spondylitis, inflammatory bowel disease and psoriasis. Safety concerns for adalimumab are similar to those for the other biologics that target TNF-α and include bacterial, fungal and viral infections, malignancies, heart failure and the development of lupus- or multiple sclerosis-like syndromes. In adults with rheumatoid arthritis, the presence of anti-adalimumab antibodies, found in about one quarter of treated patients, was associated with decreased efficacy and possibly increased adverse events.
Anakinra (Kineret ® ).
Anakinra is a recombinant form of the physiologic human protein interleukin-1 receptor antagonist (IL-1Ra). It is structurally modified from the natural molecule, in that it lacks glycosylation and it has a methionine residue added to the amino terminus. It binds the IL-1 type 1 receptor without causing signaling and thereby prevents activation by the agonistic ligands IL-1α and IL-1β. In 2001, it was approved for treatment of adults with rheumatoid arthritis. Subsequently, based on the remarkable levels of serum IL-1 found in cryopyrin-associated autoinflammatory syndromes (CAPS), anakinra was tested in an open-label uncontrolled trial of 43 neonatal-onset multisystem inflammatory disease (NOMID) patients of varying ages. All patients improved, and a subset relapsed upon withdrawal of the medication. Both the acute attacks and the progression of irreversible organ damage were inhibited. These findings led to the approval of NOMID as an additional indication for anakinra in 2014. Although anakinra has shown some evidence for efficacy in systemic juvenile idiopathic arthritis (SJIA), and is used in practice for this condition, it does not have FDA approval for this indication. Anakinra has a relatively short in vivo half-life (4–6 hours), and must be given daily by subcutaneous injection. The main safety concern has been an increased susceptibility to infection.
Basiliximab (Simulect ® ).
Basiliximab is a mouse/human chimeric monoclonal antibody specific for the α chain of the high-affinity IL-2 receptor (CD25). By blocking the cytokine IL-2 from binding to its receptor, basiliximab inhibits the activation of T lymphocytes. It is therefore effective for the prevention of acute rejection after renal transplantation. It was approved for this use in adults in 1998. Although only open-label studies have been documented in pediatric patients, an explicit indication for use in this population was added by the FDA in 2001. It is approved for use with an immunosuppressive regimen including cyclosporine and corticosteroids. It is administered as a single intravenous dose 2 hours before transplantation, and a second dose 4 days later. Infections are a prominent complication in this highly immunosuppressed patient group; however, the contribution of basiliximab per se to these adverse events is not clear. Anaphylaxis-like immediate hypersensitivity reactions have been reported in post-marketing surveillance.
Canakinumab (Ilaris ® ).
Canakinumab is a fully human recombinant monoclonal antibody specific for IL-1β. It therefore blocks the binding of this cytokine to its receptor. It received a priority review approval in 2009 for use in two CAPS – familial cold autoinflammatory syndrome (FCAS) and Muckle-Wells syndrome (MWS) – in patients ≥ 4 years of age. Supporting data came from an international double-blind, placebo controlled 24-week withdrawal study of 35 patients, and from uncontrolled experience with an additional 69 patients (including 23 pediatric patients overall). Strikingly, 97% of patients in the controlled study had a complete response by day 29 of the open-label treatment phase. During the withdrawal period, none of the active drug-treated patients relapsed, while 81% in the placebo arm had a disease flare. In 2013, canakinumab received approval for the additional indication of SJIA in children ≥ 2 years of age, based on a 29-day double-blind, placebo-controlled trial of 84 patients, and a double-blind, placebo-controlled withdrawal trial of 100 patients over up to two years of treatment. Although these trials indicated clinical efficacy, the magnitude of the effect was not nearly as impressive as for the CAPS trials. Canakinumab is administered subcutaneously every 8 weeks for CAPS and every 4 weeks for SJIA. The most important safety concern has been serious infections.
Denosumab (Xgeva ® ).
Denosumab is a fully human recombinant monoclonal antibody specific for RANKL (receptor activator of NF-κB ligand). It blocks the binding of this ligand with RANK on pre-osteoclasts and thereby prevents their maturation into osteoclasts. It thus favors bone formation over bone resorption. Animal studies have indicated that denosumab can interfere with bone development, so its pediatric use, approved in 2013, has been restricted to skeletally mature adolescents with giant cell tumors of the bone. Two open-label, uncontrolled trials provided evidence of efficacy. In the first trial in 37 adults, 30 were considered to have responded by either histologic or radiologic criteria. In a second trial of 282 patients, including 10 adolescents (13–17 years old), almost no disease progression was seen with a median follow-up of 9 to 13 months, and about half the patients were considered to have a complete or partial tumor response (an exploratory outcome in this phase II study). However, an independent review of the data in the two trials concluded that only 25% of patients, including two of the six evaluable adolescents, showed a partial response (Xgeva® package insert). For treatment of giant cell tumors, denosumab is administered subcutaneously on days 1, 8, 15 and 30, and then monthly. The major safety concerns have been hypocalcemia and osteonecrosis. Note that denosumab is also marketed for treatment of osteopenia/osteoporosis under a separate brand name (Prolia®), without a pediatric indication.
Ecallantide (Kalbitor ® ).
Ecallantide is a 7,000 kDa protein that binds to the active site of kallikrein and thereby blocks the conversion of high molecular weight kininogen to bradykinin. Its structure is based on the first Kunitz domain (active site) of human tissue factor inhibitor (also known as lipoprotein associate coagulation inhibitor). It was selected by phage display to bind kallikrein with high affinity, and it is produced in yeast by recombinant technology. It was approved in 2009 for the treatment of acute episodes of hereditary angioedema (HAE) due to C1 esterase deficiency or dysfunction, based on two randomized, double-blind controlled trials in a total of 143 HAE patients ≥ 10 years old. Not surprisingly, most of the enrolled patients in these trials were adults, and the pediatric population was skewed toward the older ages. Thus, the original labeling was for patients ≥ 16 years old; in 2014, the FDA extended the labeling to include adolescents ≥ 12 years old. The pivotal studies, which were single dose, did not report drug-related serious adverse events, but the package labeling, which includes experience in retreated patients, cites a 4% incidence of anaphylaxis. This complication has not been specifically seen in the published pediatric experience, but the number of individuals < 18 years old analyzed is relatively small ( N = 29).
Eculizumab (Soliris ® ).
Eculizumab is a humanized recombinant monoclonal antibody that binds the C5 complement component and prevents its activation by cleavage. This blocks the release of the inflammatory C5a fragment and prevents the subsequent initiation of the terminal complement cascade (C5b–C9). It was approved in 2007 for the treatment of adults with paroxysmal nocturnal hemoglobinuria (PNH). In 2009, eculizumab received an accelerated approval for treatment of atypical hemolytic uremic syndrome (aHUS) on the basis of two prospective, open-label, uncontrolled trials with a total of 37 treated patients, of which 6 were adolescents, and a retrospective experience with 17 pediatric patients age 2 months to 17 years (unpublished, summarized in the package insert). Most patients in all the studies showed improvement in blood counts and renal function, and freedom from need for plasma therapy. It is not known if eculizumab will be effective in infection-induced HUS. Treatment is by intravenous administration, initially every week and then every 2 weeks, continued indefinitely. Whether therapy can be stopped successfully in some patients is subject to further investigation, as is the potential use of eculizumab in children with PNH. In analogy with the congenital terminal component complement deficiencies, eculizumab therapy is associated with increased susceptibility to infections with Gram-negative organisms, including Neisseria meningitidis .
Etanercept (Enbrel ® ).
Etanercept is a soluble form of the p75 receptor for TNF. It is a recombinant chimeric homodimer consisting of two polypeptide chains that splice the TNF receptor subunit to the Fc portion of IgG1. It binds both TNF-α and TNF-β (also known as lymphotoxin alpha, LTα) in both the soluble and transmembrane forms. It was first approved for the treatment of adult rheumatoid arthritis in 1998. In 1999, the indication for polyarticular JIA in patients ≥ 4 years old was added on the basis of an open-label study of 69 patients, followed by a controlled, double-blind 4-month withdrawal study on the 51 patients who initially responded. In 2007, the age indication was lowered to ≥ 2 years on the basis of open-label unpublished experience (mentioned in the package insert). Etanercept is administered by subcutaneous injection 1 to 2 times per week. Safety concerns listed in the package insert include infections, malignancies (particularly lymphomas) and the development of autoimmune disease such as multiple sclerosis or lupus. Etanercept therapy may have a lower risk for activation of tuberculosis, compared to monoclonal antibodies against TNF. In addition, the association of malignancies with etanercept and other anti-TNF agents has been questioned in recent analyses. Etanercept also has adult indications for ankylosing spondylitis, psoriasis and psoriatic arthritis. It does not have the same effectiveness in inflammatory bowel disease as the monoclonal antibody anti-TNF agents.
Infliximab (Remicade ® ).
Infliximab was the first anti-TNF monoclonal antibody to be approved, starting in 1998 with the indication for Crohn’s disease in adults. Adult rheumatoid arthritis was included in 1999 and adult ulcerative colitis in 2005. Treatment of children (≥ 6 years old) with Crohn’s disease was added to the label in 2006, based on an open-label trial of an initial 10-week treatment period, followed by randomization of responding patients to maintenance treatment every 8 weeks or every 12 weeks. Pediatric ulcerative colitis received approval in 2011 based on a very similar open-label study. A phase III trial of 122 children (ages 4–17) with polyarticular JIA failed to meet its primary outcome for statistically significant efficacy at 14 weeks, so this indication does not appear on the label. Nevertheless, the general anecdotal experience has been positive, and infliximab is included among the TNF inhibitors recommended by the American College of Rheumatology for treatment of JIA. Childhood autoimmune uveitis is another relatively common off-label usage. Safety issues with infliximab include infections, especially tuberculosis. A relatively high incidence of immune responses to infliximab (HACA) has been seen in children, as have serious infusion reactions. Infliximab is administered intravenously at weeks 0, 2 and 6, and then every 8 weeks.
Omalizumab (Xolair ® ).
Omalizumab is a recombinant humanized monoclonal antibody that binds the constant region of free IgE. It thus blocks the binding of IgE to cell surface receptors on basophils and mast cells, but it does not cross-link IgE that is already cell bound. It causes a large increase in serum IgE, in the form of omalizumab-IgE complexes. It was approved in 2003 for the treatment of severe to moderate chronic allergic asthma in patients ≥ 12 years old who are not controlled with inhaled corticosteroids. It is not used in the setting of acute exacerbations. Two other trials evaluated omalizumab in children between 6 and 12 years old. Although the one efficacy trial met its primary endpoint (rate of asthma exacerbations), other efficacy outcomes did not show statistical superiority over placebo. Based on safety concerns (see below), it was decided that it was not appropriate to extend the label indication to children < 12 years old. In 2014, the indication for chronic idiopathic urticaria was added with the same age limitation. Data to support this label change came from two phase III studies with a total of 640 patients treated over 12 or 24 weeks. The major safety issues with omalizumab have been anaphylaxis, serum sickness, parasitic infestations and malignancy. Omalizumab is administered subcutaneously every 2 to 4 weeks.
Palivizumab (Synagis ® ).
Palivizumab is a humanized monoclonal antibody specific for the envelope fusion protein (RSV-F) of respiratory syncytial virus. It thus prevents cell entry by the virus and cell-to-cell fusion of RSV-infected cells. Its approval in 1998 was based on demonstration of its efficacy in preventing serious RSV infection in two double-blind, randomized, placebo-controlled trials with 2,789 children ≤ 2 years of age who were considered to be at high risk because of congenital heart disease, bronchopulmonary dysplasia or prematurity. Palivizumab was given intramuscularly in five monthly injections beginning prior to the RSV season. The incidence of hospitalization with proven RSV infection was decreased about 50% in both studies. Children who were hospitalized for RSV infection despite having received the active drug did not show a milder course of disease. The package insert states that palivizumab is indicated for prophylaxis against serious lower respiratory tract RSV in high-risk children. The major safety concern has been anaphylaxis (rare) and other hypersensitivity reactions. Palivizumab is not indicated for the treatment of RSV infection.
Raxibacumab.
Raxibacumab is a fully human monoclonal antibody that is directed at the protective antigen (PA) moiety of the anthrax toxin (from Bacillus anthracis ) and that prevents the toxin from entering cells. It is indicated for the treatment of inhalation anthrax, in combination with appropriate antibiotics. It is also indicated for anthrax prophylaxis if alternatives are not feasible. It is administered as a single intravenous dose. Its approval in 2012 was based on its low toxicity profile in over 500 normal (adult) volunteers, and therapeutic efficacy in monkey and rabbit models of anthrax. For evident ethical and logistical issues, it has never been tested in humans for its approved indication. It is available only from the Centers for Disease Control and Prevention(CDC).
Rilonacept (Arcalyst ® ).
Rilonacept is a genetically engineered cytokine ‘trap’ that binds IL-1β with very high affinity. With lower affinity, it also binds IL-1α and IL-1Ra. Rilonacept was approved in 2008 for treatment in patients ≥ 12 years old with CAPS. Approval was based on two sequential randomized controlled trials with the same 47 adult patients (44 with FCAS; 3 with MWS): part A consisted of a 6-week placebo-controlled treatment period; this was immediately followed by part B, which began with 9 weeks of patient-blinded active treatment, followed by 9 weeks of placebo-controlled randomized withdrawal. The institution of rilonacept produced a rapid and sustained reduction in patient-reported symptoms (the primary outcome) in the great majority of subjects, while the group withdrawn to placebo experienced a return in symptoms beginning at around 3 weeks. Treatment also normalized serum markers of inflammation, including CRP and serum amyloid protein. Pediatric data were not required with the initial approval, as rilonacept was given an orphan drug designation. Clinical responses were maintained over a subsequent 72-week open-label extension study of 44 subjects from the pivotal trial plus 57 new patients. The additional patients included eight children, age 12 to 17. Two deaths in adult patients were caused by pneumococcal meningitis and coronary artery disease, respectively, but were not considered to be drug related. The most common drug-related adverse event has been injection site reactions. About one quarter of treated patients have been found to develop anti-rilonacept antibodies, usually in low titer. In two patients this was associated with pharmacokinetic changes, but the clinical significance of these findings is not yet apparent. Rilonacept is administered by weekly subcutaneous injections.
Tocilizumab (Actemra ® ).
Tocilizumab is a recombinant humanized monoclonal antibody that binds the soluble and membrane-bound forms of the IL-6 receptor and prevents its interaction with IL-6. It was approved for the treatment of adult rheumatoid arthritis in 2010. An indication for systemic JIA was added in 2011, and for polyarticular JIA in 2013. Supporting data for the SJIA label came from a 12-week randomized, placebo-controlled trial of 112 children ages 2 to 17, in which the tocilizumab-treated patients showed substantially more improvement than the placebo group (e.g. ACR70 of 71% vs 8%). Improvement was maintained over a 40-week open-label extension study. Supporting data for the polyarticular JIA label came from a three-phase study of 188 patients ages 2 to 17. An active 16-week lead-in period resulted in 166 patients achieving an ACR30 and progressing to the 40-week randomized, placebo-controlled withdrawal period with disease flare as a primary outcome measure. Twenty-six percent of tocilizumab-treated patients experienced a flare, compared to 48% of placebo-treated patients. Responses were maintained over a further 48-week open-label extension. Significant adverse events have included neutropenia, elevated cholesterol and liver function tests, and serious infections. Macrophage activation syndrome was seen in some of the clinical trials, with an overall incidence of one to two per 100 patient years, which was not felt to represent an increased risk. Tocilizumab is administered to children in the intravenous preparation, every 4 weeks for polyarticular JIA and every 2 weeks for systemic JIA.