You are on page 1of 12

Regulatory Toxicology and Pharmacology 73 (2015) 595e606

Contents lists available at ScienceDirect

Regulatory Toxicology and Pharmacology


journal homepage: www.elsevier.com/locate/yrtph

Proof of concept for a banding scheme to support risk assessments


related to multi-product biologics manufacturing
Jeffrey W. Card a, *, Hana Fikree a, Lois A. Haighton a, James Blackwell b, Brian Felice c,
Teresa L. Wright d
a
Intertek Scientific & Regulatory Consultancy, Mississauga, ON, Canada
b
The Quantic Group, Livingston, NJ, USA
c
Shire, Lexington, MA, USA
d
Dimension Therapeutics, Inc., Cambridge, MA, USA

a r t i c l e i n f o a b s t r a c t

Article history: A banding scheme theory has been proposed to assess the potency/toxicity of biologics and assist with
Received 26 June 2015 decisions regarding the introduction of new biologic products into existing manufacturing facilities. The
Received in revised form current work was conducted to provide a practical example of how this scheme could be applied. Infor-
31 August 2015
mation was identified for representatives from the following four proposed bands: Band A (lethal toxins);
Accepted 2 September 2015
Band B (toxins and apoptosis signals); Band C (cytokines and growth factors); and Band D (antibodies,
Available online 8 September 2015
antibody fragments, scaffold molecules, and insulins). The potency/toxicity of the representative sub-
stances was confirmed as follows: Band A, low nanogram quantities exert lethal effects; Band B, repeated
Keywords:
Biologics
administration of microgram quantities is tolerated in humans; Band C, endogenous substances and re-
Manufacturing combinant versions administered to patients in low (interferons), intermediate (growth factors), and high
Cross-contamination (interleukins) microgram doses, often on a chronic basis; and Band D, endogenous substances present or
Cleaning validation produced in the body in milligram quantities per day (insulin, collagen) or protein therapeutics adminis-
Impurities tered in milligram quantities per dose (mAbs). This work confirms that substances in Bands A, B, C, and D
Risk assessment represent very high, high, medium, and low concern with regard to risk of cross-contamination in
manufacturing facilities, thus supporting the proposed banding scheme.
© 2015 Elsevier Inc. All rights reserved.

1. Introduction pharmacological effects, since a desired pharmacological effect in


an intended recipient may be undesirable or dangerous in an un-
It is widely recognized that manufacturing multiple pharma- intended recipient.
ceutical products in a single facility is associated with risks related Several guidance documents and thought papers have been
to cross-contamination. Carryover of a given drug substance [or published on the topics of cross-contamination of APIs and rec-
active pharmaceutical ingredient (API)] into another drug product ommended methods to establish safe exposure limits that can be
represents a potential risk for cross-contamination that can be of used to assist with cleaning validation efforts (Dolan et al., 2005;
concern to a patient who is subsequently administered the ISPE, 2010; Walsh, 2011a,b,c; Bercu et al., 2013; Sargent et al.,
contaminated drug product. This risk can be due to known toxi- 2013). The derived exposure limit for a given substance is typi-
cological effects of the contaminating substance or to its recognized cally referred to as a permitted daily exposure (PDE) value or an
acceptable daily exposure (ADE) value that is applicable for lifetime
daily exposure in all patient populations and by all routes of
Abbreviations: ADE, acceptable daily exposure; API, active pharmaceutical
ingredient; FDA, Food and Drug Administration; G-CSF, granulocyte colony stimu- exposure. From a regulatory perspective, it is noted that the Euro-
lating factor; GM-CSF, granulocyte-macrophage colony stimulating factor; IFN, pean Medicines Agency (EMA) has issued a guideline on estab-
interferon; IL, interleukin; IM, intramuscular; IU, international unit; IV, intravenous; lishing health based exposure limits (PDE values) related to
LD50, median lethal dose; mAbs, monoclonal antibodies; MTD, maximum tolerated
manufacturing different medicinal products in shared facilities
dose; PDE, permitted daily exposure; SC, subcutaneous; TNF, tumor necrosis factor.
* Corresponding author. 2233 Argentia Road, Suite 201, Mississauga, ON, L5N 2X7,
which came into effect in June 2015 (EMA, 2014).
Canada. The existing literature and guidance documents focus on small
E-mail address: jeffrey.card@intertek.com (J.W. Card).

http://dx.doi.org/10.1016/j.yrtph.2015.09.003
0273-2300/© 2015 Elsevier Inc. All rights reserved.
596 J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606

molecule APIs, and very little attention appears to have been given Clostridium botulinum. There are four naturally occurring botulism
to large molecule biologics. One notable exception is the article by syndromes, namely foodborne botulism, wound botulism, infant
Mott et al. (2013) which describes several methods that can be used botulism (caused by intestinal colonization and toxin production),
to evaluate the extent of biopharmaceutical product inactivation as and adult intestinal toxemia (caused by intestinal colonization and
it relates to cleaning validation and potential carryover on toxin production). All forms of botulism have the same clinical
manufacturing equipment. In this regard, the EMA guideline notes syndrome comprised of cranial nerve paralysis followed by
that most cleaning processes employ extremes of pH and/or heat descending symmetrical flaccid paralysis of voluntary muscles; in
and that therapeutic macromolecules and peptides are “known to the worst cases this can progress to respiratory compromise and
degrade and denature when exposed to pH extremes and/or heat, death (Sobel, 2005).
and may become pharmacologically inactive” (EMA, 2014). As such, There are seven immunologically distinct botulinum toxins,
the guideline concludes that it may not be necessary to use a PDE designated by the letters A to G, which are responsible for causing
approach to determine health based exposure limits for biologics. botulism. Botulinum toxins A, B, E, and F cause human forms of
This language is somewhat ambiguous, and concerns may remain in botulism while toxins C and D cause botulism in animals; botuli-
cases where cleaning efforts are insufficient. In addition, knowledge num toxin G has not been reported to cause botulism in humans
of potential exposure limits for biologics or of the potency/toxicity of (Kotsonis and Burdock, 2008). All of the botulinum toxins are
one type of biologic relative to that of another would assist in early neurotoxic. Their mechanism of action involves blockade of
decisions regarding the suitability of introducing a new biologic into acetylcholine release from presynaptic motor-neuron terminals
a facility in which other biologics are already manufactured. thereby preventing stimulation of motor fibers. Recovery from
A banding scheme for assessing the potency/toxicity of biologics botulism often takes weeks to months, and respiratory distress and
was proposed in a recent article by Carver (2013). This concept paralysis may persist for six to eight months (Kotsonis and Burdock,
would involve categorizing biologics into different “bands” based 2008).
on their potency and toxicological potential. This banding scheme is Botulinum toxins are some of the most potent toxins known.
intended to help identify more potent/toxic biologics from those Data summarized by Gill (1982) demonstrate the potency of bot-
that are more innocuous, and is anticipated to assist with decisions ulinum toxins in mice, with reported median lethal dose (LD50)
regarding bringing new products into an existing facility or values ranging from 0.4 ng/kg body weight for botulinum toxin D
whether to outsource manufacturing efforts to a dedicated facility (intraperitoneal) to 2.5 ng/kg body weight for botulinum toxin F
and therefore eliminate the potential for cross-contamination. As [intravenous (IV)]. As noted by Sobel (2005), commonly cited
outlined by Carver (2013), the proposed banding scheme and ex- estimated lethal doses for botulinum toxin A for a 70 kg human are
amples of substances within each band are as follows (potency/ 90e150 ng when introduced intravenously (approximately
toxicity decreases from Band A to Band D): 1.3e2.1 ng/kg body weight), 800e900 ng when introduced via
inhalation, and 70 mg (70,000 ng) when introduced orally. Others
 Band A: Lethal toxins (e.g., botulinum toxin); have estimated that the lethal dose of botulinum toxin in humans
 Band B: Toxins and apoptosis signals [e.g., tumor necrosis factor- may be as low as 1 ng (Kotsonis and Burdock, 2008).
a (TNF-a)];
 Band C: Cytokines, growth factors, etc. (e.g., interferons, in- 3.1.2. Diphtheria toxin
terleukins, growth factors); and Diphtheria is an acute, communicable disease caused by the
 Band D: Antibodies, antibody fragments, scaffold molecules, and exotoxin-producing bacteria Corynebacterium diphtheria (Hadfield
insulins [e.g., insulins, monoclonal antibodies (mAbs), antibody et al., 2000). It is normally transmitted by sneezing, coughing, or
fragments, scaffold proteins]. direct contact. The bacteria usually localizes in the upper respira-
tory tract where it induces the formation of an inflammatory
While the proposed banding scheme appears to make sense pseudomembrane. From here, released diphtheria toxin is absor-
intuitively, quantitative or qualitative information are not provided bed into the systemic circulation and causes severe systemic pa-
to support the proposed bands or the example substances that are thology. Diphtheria toxin does not have a specific target organ
indicated within each band. The purpose of the work summarized although peripheral nerves and myocardium appear to be most
here was to determine whether there is quantitative and/or quali- often affected (Hadfield et al., 2000). Diphtheria toxin enters cells
tative information to support the proposed bands from a scientific via binding to heparin binding epidermal growth factor precursor
perspective. Such information would be useful to consider when that is present on cell membranes. Once inside the cell, diphtheria
conducting risk assessments of biologics pertaining to cleaning toxin acts to inhibit protein synthesis and ultimately leads to pro-
validation and cross-contamination. grammed cell death (Shapira and Benhar, 2010).
Diphtheria toxin is extremely potent. It has been demonstrated
2. Methods that introduction of one molecule of diphtheria toxin fragment A
(the catalytic domain) into the cytosol of a cell is sufficient to induce
To determine whether the proposed banding scheme could be cell death (Yamaizumi et al., 1978). Barksdale (1970) reported that
affirmed with scientific data, representative substances were diphtheria toxin is lethal to humans and animals at a dose level of
selected from each of the four proposed bands and searches for 130 ng/kg body weight; further details were not reported. A
information were performed. A summary of the representative comparably low IV LD50 value of 10 ng/kg body weight (0.01 mg/kg
substances and the resources that were searched for potency/ body weight) for diphtheria toxin in mice is presented in a publicly-
toxicity information is presented in Table 1. available database maintained by the University of Florida (avail-
able at: http://www.ehs.ufl.edu/programs/bio/toxins/toxin-table/;
3. Results last accessed April 21, 2015).

3.1. Information on representative substances from Band A 3.2. Information on representative substances from Band B

3.1.1. Botulinum toxin 3.2.1. Crotoxin


Botulism is a rare disease caused by the preformed toxins of Phospholipase A2 catalyzes the hydrolysis of phospholipids to
J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606 597

Table 1
Representative substances and sources of information.

Banda Representative substancea Information source

A Botulinum toxin (lethal toxin) Reference textbooks and the published scientific literature.
Diphtheria toxin (lethal toxin) Published scientific literature.
B Crotoxin (a less potent toxin) and TNF-a Published scientific literature.
(a pro-apoptotic factor)
C Interferons, interleukins, and growth Product monographs obtained via the Drugs@FDA
factors (representative examples from each) database (http://www.accessdata.fda.gov/scripts/cder/drugsatfda/).
D Insulin Reference textbooks, published scientific literature, and product monographs obtained
via the Drugs@FDA database (http://www.accessdata.fda.gov/scripts/cder/drugsatfda/).
Collagen Published scientific literature.
mAbs (representative examples) Reference textbooks, published scientific literature, and product monographs obtained
via the Drugs@FDA database (http://www.accessdata.fda.gov/scripts/cder/drugsatfda/).
a
As outlined by Carver (2013).

form free arachidonic acid and lysophospholipids. Numerous Acute and chronic systemic inflammatory reactions are modulated
phospholipase A2 enzymes have been identified in various snake by TNF-a, which induces its own secretion and the secretion of
venoms and appear to contribute to their effect on the host several other inflammatory cytokines and chemokines. Evidence is
(Watkins, 2008). Crotoxin is found in the venom of the South available to support a central role for TNF-a in several autoimmune
American snake, Crotalus durissus terrificus. It is a non-covalent diseases such as rheumatoid arthritis, Crohn's disease, ulcerative
complex composed of two subunits; an acidic subunit A and a colitis, multiple sclerosis, and others, and TNF-a appears to be an
basic subunit B (Cardoso et al., 2001; Cura et al., 2002). Subunit B is important risk factor for tumorigenesis, tumor progression, and
a cytotoxic and neurotoxic phospholipase A2 that is released from metastasis (Chu, 2013).
the crotoxin complex upon binding to the cell surface. TNF-a is produced and secreted mainly by macrophages
A limited number of toxic effects are induced by crotoxin, and it although it can be generated by other cell types. Based on data
is believed that the epidermal growth factor receptor plays a role in presented in 18 different human studies and summarized in the
its cell targeting (Cura et al., 2002). Neurotoxicity manifested as Plasma Proteome Database, endogenous plasma/serum concen-
peripheral blockade of neuromuscular transmission is the principal trations of TNF-a are generally very low, specifically in the low pg/
effect mediated by crotoxin, while intramuscular (IM) administra- mL range (Nanjappa et al., 2014).
tion of crotoxin has been demonstrated to cause myotoxicity (Cura A single IV dose of recombinant human TNF-a to mice was not
et al., 2002). As reported in a publicly-available database main- lethal at a dose level of up to 0.4 mg/kg body weight (400 mg/kg
tained by the University of Florida (available at: http://www.ehs.ufl. body weight) (Myers et al., 1990).
edu/programs/bio/toxins/toxin-table/; last accessed April 21, 2015), A recent review by Roberts et al. (2011) summarized data from
the IV LD50 value for crotoxin in mice is 82 mg/kg body weight. 18 Phase 1 studies and ten Phase 2 studies in cancer patients in
Crotoxin has been evaluated in a Phase 1 study in patients with which recombinant human TNF-a was administered as a single
advanced cancer (Cura et al., 2002). In this study, crotoxin was agent. All of the identified clinical trials were conducted in the
administered by IM injection for 30 consecutive days at a dose level 1980s and 1990s and all but one of the studies utilized the IV route
of 0.03, 0.06, 0.12, 0.18, 0.21, or 0.22 mg/m2/day (equivalent to 0.8, of administration (one Phase 1 study utilized IM injection). The
1.6, 3.2, 4.9, 5.7, or 5.9 mg/kg body weight/day). Pharmacokinetic design of the Phase 1 studies varied in terms of dosing posology;
data reported for the 0.21 mg/m2 (5.7 mg/kg body weight) dose level single dose administration, multiple dosing (daily to every three
on Study Days 1 and 15 indicated rapid absorption of crotoxin into weeks), and continuous IV infusion (one to five days in duration)
the bloodstream, with maximum plasma concentrations observed were evaluated in various studies. As summarized by Roberts et al.
within 20 min of dosing. No neurological toxicity was observed at (2011), a dose level of 150e200 mg/m2 administered as a 30-
dose levels 0.12 mg/m2 (3.2 mg/kg body weight). The most min infusion was identified as the MTD in several of the Phase 1
common neurological adverse effect at dose levels 0.18 mg/m2 studies. This is equivalent to a dose of 4e5.4 mg/kg body weight for
(4.9 mg/kg body weight) was diplopia (double vision). This was a 50 kg adult. Dose-limiting toxicities appeared to be generally
attributed to paresis of the external ocular muscles and was not well-tolerated and reversible; these included hypotension, throm-
observed in any patient beyond Day 21. The only other neurological bocytopenia, leucopenia, neurotoxicity, fever, nausea/vomiting, and
toxicities observed at the highest dose level (0.22 mg/m2, or 5.9 mg/ general symptoms of malaise and weakness. A total of ten Phase 2
kg body weight) were occasional incidences of ptosis, nystagmus, studies were identified in which recombinant human TNF-a was
and anxiety. No effects of crotoxin were observed on hematopoietic, administered by the IV route to patients with advanced/metastatic
hepatic, or renal function. Diarrhea and excessive salivation were cancers of various types. The most common dosing posology
occasionally observed, and one patient had an anaphylactic reac- involved a 30-min infusion of a dose of 150e200 mg/m2 (equivalent
tion on the last day of dosing. Based on the available data, the to a dose of 4e5.4 mg/kg body weight for a 50 kg adult) adminis-
maximum tolerated dose (MTD) was determined to be 0.21 mg/m2 tered daily for three to five days and repeated every one to four
(5.7 mg/kg body weight) (Cura et al., 2002). weeks. The most serious toxicities included respiratory failure and
coagulopathies, while more common adverse events included hy-
3.2.2. Tumor necrosis factor-a potension, thrombocytopenia, leucopenia, fever/chills, headache,
TNF-a is a member of the TNF family of cytokines which in- and nausea/vomiting. Notably, tumor responses were rare and
cludes TNF-b, CD40 ligand, Fas ligand (FasL), TNF-related apoptosis when they occurred, only partial responses were observed.
inducing ligand (TRAIL), and others (Chu, 2013). TNF-a and other The collective data from the available clinical studies indicates
members of the TNF family have important functions in various that systemic administration of recombinant human TNF-a has
physiological processes. TNF-a is an endogenous pyrogen that is been evaluated in several studies in cancer patients at dose levels
best known as an inducer of necrosis and apoptosis of tumor cells. generally in the range of 4e5.4 mg/kg body weight. A common
598 J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606

profile of adverse events was noted in these studies and included 0.5 m2 the recommended dose of ACTIMMUNE® is 1.5 mg/kg body
hypotension, thrombocytopenia, leucopenia, fever, and nausea/ weight three times per week administered by SC injection.
vomiting. According to the product label, the most common adverse re-
actions associated with ACTIMMUNE® include fever, headache,
3.3. Information on representative substances from Band C rash, chills, injection site reactions, fatigue, and others. Other
adverse reactions include decreased mental status, gait distur-
3.3.1. Interferons bance, dizziness, bone marrow toxicity (reversible neutropenia and
3.3.1.1. Interferon alpha-2b. Interferon alpha-2b (IFNa-2b) is an thrombocytopenia), and hepatic toxicity.
endogenous cytokine that induces the innate anti-viral immune
response. Its biological effects include inhibition of viral replication 3.3.2. Interleukins
in virus-infected cells, the suppression of cell cycle progression/cell 3.3.2.1. Interleukin-2. Interleukin-2 (IL-2) is an endogenous cyto-
proliferation, induction of apoptosis, anti-angiogenic activities, and kine that has a number of immunoregulatory properties, including
numerous immunomodulating activities, such as enhancement of the following:
the phagocytic activity of macrophages, activation of NK cells,
stimulation of cytotoxic T-lymphocytes, and the upregulation of the  Enhancement of lymphocyte mitogenesis and stimulation of
Th1 T-helper cell subset. long-term growth of human IL-2-dependent cell lines;
PegIntron® (Merck & Co., Inc.) is a covalent conjugate of re-  Enhancement of lymphocyte cytotoxicity;
combinant human IFNa-2b and polyethylene glycol. The pegylation  Induction of killer cell [lymphokine-activated (LAK) and natural
of IFNa-2b allows for a longer circulation time, permitting a (NK)] activity; and
decreased dosing frequency relative to free IFNa-2b. PegIntron® is  Induction of IFNg production.
indicated for the treatment of chronic Hepatitis C in patients with
compensated liver disease. The recommended dose of PegIntron® is Proleukin® (Prometheus Laboratories Inc.) is a recombinant
1.5 mg/kg body weight/week by subcutaneous (SC) injection for one human IL-2 that differs from native IL-2 in the following ways: i) it
year. is not glycosylated because it is derived from Escherichia coli; ii) it
According to the product label, the most common adverse re- has no N-terminal alanine as the codon for this amino acid was
actions (>40%) in adult patients receiving PegIntron® are injection deleted during the genetic engineering procedure; and iii) it has
site inflammation/reaction, fatigue/asthenia, headache, rigors, fe- serine substituted for cysteine at amino acid position 125 (accom-
vers, nausea, myalgia and anxiety/emotional lability/irritability. The plished by site-specific manipulation during the genetic engineer-
most common adverse reactions (>25%) in pediatric patients ing procedure). Proleukin® is indicated for treatment of adults with
receiving PegIntron® are pyrexia, headache, neutropenia, fatigue, renal cell carcinoma or metastatic melanoma. The recommended
anorexia, injection-site erythema, and vomiting. dose of Proleukin® is 0.037 mg/kg body weight (37 mg/kg body
weight) every eight hours by 15-min IV infusion for a total of 14
3.3.1.2. Interferon beta-1a. Interferon beta-1a (IFNb-1a) is an doses. This is repeated after nine days of rest for a total of 28 doses
endogenous cytokine that is produced by eukaryotic cells in per course, as tolerated.
response to viral infection and other biologic agents. IFNb-1a pos- A boxed warning on the product label for Proleukin® indicates
sesses immunomodulatory, anti-viral, and anti-proliferative bio- that it has been associated with “capillary leak syndrome, which is
logical activities. characterized by a loss of vascular tone and extravasation of plasma
Avonex® (Biogen Idec Inc.) and Rebif® (EMD Serono, Inc.) are proteins and fluid into the extravascular space”. Several other
products containing recombinant IFNb-1a that are indicated for the adverse effects are listed in the product label.
treatment of relapsing forms of multiple sclerosis to slow the
accumulation of physical disability and decrease the frequency of 3.3.2.2. Interleukin-11. Interleukin-11 (IL-11) is an endogenous
clinical exacerbations. The recommended dose of Avonex® is 30 mg cytokine that stimulates proliferation of hematopoietic stem cells
once per week administered by IM injection, while the recom- and megakaryocyte progenitor cells and induces megakaryocyte
mended dose of Rebif® is 22 or 44 mg three times per week maturation resulting in increased platelet production.
administered by SC injection. Neumega® (Wyeth Pharmaceuticals, Inc.) is a recombinant hu-
According to the product labels for Avonex® and Rebif®, the man IL-11 that differs from native IL-11 only in that it lacks the
most common adverse reactions are flu-like symptoms including amino-terminal proline residue. Neumega® is indicated for the
chills, fever, myalgia, and asthenia. Other, more severe adverse re- prevention of severe thrombocytopenia and the reduction of the
actions include depression, suicidal thoughts, and psychotic dis- need for platelet transfusions following myelosuppressive chemo-
orders, hepatic injury, anaphylaxis or other allergic reactions, therapy in adult patients with non-myeloid malignancies who are
congestive heart failure, decreased peripheral blood counts, sei- at high risk of severe thrombocytopenia. The recommended dose of
zures, and autoimmune disorders. Neumega® is 0.05 mg/kg body weight/day (50 mg/kg body weight/
day) administered as a SC injection for a maximum of 21 days per
3.3.1.3. Interferon gamma-1b. Interferon gamma-1b (IFNg-1b) is an treatment course. Treatment should be initiated six to 24 h after
endogenous cytokine that is produced by various immune cells. It is completion of chemotherapy and continued until the post-nadir
an activator of macrophages and has anti-viral, immunoregulatory, blood platelet count is >50,000/mL.
and anti-tumor properties. A number of adverse effects are listed in the Neumega® product
ACTIMMUNE® (InterMune, Inc.) is recombinant human IFNg-1b label, including allergic reactions and anaphylaxis.
that is indicated for reducing the frequency and severity of serious
infections associated with Chronic Granulomatous Disease and for 3.3.3. Growth factors
delaying time to disease progression in patients with severe, ma- 3.3.3.1. Granulocyte colony stimulating factor. Granulocyte colony
lignant osteoporosis. The recommended dose of ACTIMMUNE® for stimulating factor (G-CSF) is an endogenous glycoprotein that is
patients with a body surface area >0.5 m2 is 50 mg/m2 (equivalent to produced by monocytes, fibroblasts, and endothelial cells. It stim-
approximately 1.35 mg/kg body weight) three times per week ulates proliferation, differentiation commitment, and some end-
administered by SC injection. For patients with a body surface area cell functional activation of neutrophils.
J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606 599

NEUPOGEN® (Amgen Inc.) is recombinant human G-CSF that  To prolong survival of patients who are experiencing graft fail-
differs from native G-CSF only in that it has an additional N-ter- ure or engraftment delay, in the presence or absence of infec-
minal methionine (necessary for expression in E. coli) and it is not tion, following autologous or allogeneic bone marrow
glycosylated (because it is produced in E. coli). NEUPOGEN® is transplantation.
indicated for the following:
The recommended dose of LEUKINE® is 250 mg/m2/day
 To decrease the incidence of infection, as manifested by febrile (approximately 6.75 mg/kg body weight/day) administered by IV
neutropenia, in patients with non-myeloid malignancies infusion, although the duration of treatment varies depending on
receiving myelosuppressive anti-cancer drugs associated with a the indication. For the treatment of bone marrow transplantation
significant incidence of severe neutropenia with fever; failure or engraftment delay, the recommended duration of treat-
 To reduce the time to neutrophil recovery and the duration of ment is 14 days.
fever following induction or consolidation chemotherapy According to the product label, severe adverse reactions that
treatment of adults with Acute Myeloid Leukemia; have been associated with LEUKINE® include fluid retention
 To reduce the duration of neutropenia and neutropenia-related (edema, capillary leak syndrome, and pleural and/or pericardial
clinical sequelae (e.g., febrile neutropenia) in patients with non- effusion), respiratory symptoms (dyspnea), and cardiovascular
myeloid malignancies undergoing myeloablative chemotherapy symptoms (transient supraventricular arrhythmia). Other adverse
followed by marrow transplantation; reactions include fever, headache, bone pain, chills, myalgia,
 To mobilize hematopoietic progenitor cells into the peripheral dizziness, hypotension, and injection site reactions.
blood for collection by leukapheresis; and
 To reduce the incidence and duration of sequelae of neutropenia
(e.g., fever, infections, oropharyngeal ulcers) in symptomatic 3.4. Information on representative substances from Band D
patients with congenital neutropenia, cyclic neutropenia, or
idiopathic neutropenia. 3.4.1. Insulin
The processes whereby insulin is synthesized, stored, and
The recommended dose of NEUPOGEN® varies depending on secreted by b-cells of the pancreas are well understood. Insulin
the indication. For cancer patients receiving myelosuppressive secretion is a tightly regulated process that provides for stable
chemotherapy, the recommended dose is 5 mg/kg body weight/day glucose levels in the bloodstream during periods of fasting and
for up to two weeks, administered by SC injection, short IV infusion, feeding, with glucose serving as the primary stimulus for insulin
or continuous SC or IV infusion. For patients receiving a bone secretion. The half-life of insulin in the blood is approximately five
marrow transplant, the recommended dose is 10 mg/kg body to six minutes, with degradation occurring mainly in the liver,
weight/day administered by IV or SC infusion at least 24 h after kidney, and muscle via the enzyme insulinase.
receipt of bone marrow infusion; treatment should continue until Under normal fasting conditions, insulin secretion from the
neutrophil recovery is achieved. For blood progenitor cell collec- pancreas occurs at a rate of approximately 25 ng/kg body weight/
tion, the recommended dose is 10 mg/kg body weight/day by SC minute (Hall, 2011a). This is equivalent to “background” rates of
bolus injection or continuous infusion for at least four days prior to approximately 0.25 mg/min for a 10 kg child and 1.25 mg/min for a
collection. For patients with cyclic or idiopathic neutropenia, the 50 kg adult, or 360 and 1800 mg/day, respectively. It should be noted
recommended dose is 5 mg/kg body weight/day by SC injection. that these “background” rates are based on insulin secretion only
According to the product label, severe adverse reactions asso- during fasting and do not account for the higher insulin secretion
ciated with NEUPOGEN® include allergic reactions, splenic rupture, that occurs after meals.
acute respiratory distress syndrome, and alveolar hemorrhage and Several human insulin drug products are available, including
hemoptysis (expectoration of blood). Other common adverse re- Humulin® R (Eli Lilly and Company) and Novolin® R (Novo Nordisk
actions include medullary bone pain, nausea/vomiting, alopecia, A/S). These products are intended to be administered by SC injec-
and diarrhea. tion prior to meals. Based on information provided in the product
labels, the total daily insulin requirement varies but usually ranges
3.3.3.2. Granulocyte-macrophage colony stimulating factor. from 0.5 to 1.0 units/kg body weight/day. According to the National
Granulocyte-macrophage colony stimulating factor (GM-CSF) is an Institute for Biological Standards and Control (NIBSC, 2010), one
endogenous growth factor that is secreted by various cell types. It international unit (IU) of human insulin corresponds to the activity
promotes the number and function of white blood cells, including contained in 0.03846 mg of insulin (i.e., there are 26.0 IU/mg of
neutrophils, monocytes/macrophages, and dendritic cells. human insulin). As such, the total daily insulin requirement of
LEUKINE® (Bayer HealthCare Pharmaceuticals, Inc.) is recombi- 0.5e1.0 units/kg body weight/day is equivalent to approximately
nant human GM-CSF that differs from native GM-CSF only by the 0.019e0.038 mg/kg body weight/day. For a 10 kg child this is
substitution of leucine at position 23. LEUKINE® is indicated for the equivalent to approximately 190e380 mg/day, while for a 50 kg
following: adult this is equivalent to approximately 950 to1900 mg/day. These
values are comparable to the “background” rates of insulin secre-
 To shorten time to neutrophil recovery and to reduce the inci- tion that were calculated in the preceding paragraph.
dence of severe and life-threatening infections and infections According to the product labels for Humulin® R and Novolin® R,
resulting in death after induction chemotherapy in older adults hypoglycemia is the most common adverse reaction associated
with acute myelogenous leukemia; with all insulin therapies. Excess insulin may cause hypoglycemia
 To mobilize hematopoietic progenitor cells into peripheral and hypokalemia, particularly after IV administration. Mild epi-
blood for collection by leukapheresis; sodes of hypoglycemia can usually be treated with oral glucose.
 To accelerate myeloid recovery in patients with non-Hodgkin's Severe hypoglycemia may lead to unconsciousness and/or convul-
lymphoma, acute lymphoblastic leukemia, and Hodgkin's dis- sions and may result in temporary or permanent impairment of
ease undergoing autologous bone marrow transplantation; brain function, or death. More severe episodes with coma, seizure,
 To accelerate myeloid recovery in patients undergoing alloge- or neurologic impairment may be treated with IM/SC glucagon or
neic bone marrow transplantation; and concentrated IV glucose.
600 J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606

3.4.2. Collagen immunogenicity, thus improving their overall risk-benefit ratio. In


Collagen is an endogenous compound produced primarily by general, mAbs are considered to be well-tolerated in humans
fibroblasts. It is insoluble in water and is composed of a triple helix despite containing elements that may be recognized by the re-
which contains a high content of hydroxyproline. Collagen is a cipient's immune system as being foreign (Hansel et al., 2010).
major constituent of the extracellular matrix in mammals and is the A search of the Drugs@FDA database (conducted April 22, 2015)
most abundant protein in the human body (Rennie, 1999; Di Lullo using the term “mab” identified a total of 40 therapeutic mAb drug
et al., 2002; Smith and Rennie, 2007). By one estimate, collagen products that have been approved by the FDA, although not all of
comprises 3.5 kg of the weight of a 70 kg human, or approximately them are currently marketed in the U.S. Moreover, three of these
30% of total body protein (Smith and Rennie, 2007). Approximately products are actually antibodyedrug conjugates [gemtuzumab
28 types of collagen have been identified to date (Smith and Rennie, ozogamicin (Mylotarg®; Wyeth Pharmaceuticals Inc.), brentuximab
2007). Current evidence suggests that there are fast-turnover vedotin (Adcetris®; Seattle Genetics), and ado-trastuzumab
(soluble or immature) and slower-turnover (insoluble or mature) emtansine (Kadcyla™; Genentech, Inc.)]. A brief summary of
pools of collagen, with the proportion of soluble collagen converted these 40 therapeutic mAb drug products, including indications,
to insoluble collagen being dependent on the tissue and on the age dose routes, and dose levels, is provided in Table 2.
and nutritional status of the animal (Smith and Rennie, 2007). In- As noted above in Table 2, the majority of approved therapeutic
formation was not identified on the presence of collagen in blood, mAb drug products are indicated for the treatment of various types
although markers of collagen degradation (e.g., crosslinked telo- of cancer and arthritis. With the exception of palivizumab (Syna-
peptides of type I collagen) can be measured in blood and urine gis®; MedImmune, LLC) and ranibizumab (Lucentis®; Genentech,
(Smith and Rennie, 2007). Inc.), which are administered by the IM and intravitreal routes,
Collagen is a major constituent of intercellular connective tissue, respectively, all of the therapeutic mAbs that have been approved
including that of meats that are ingested by humans. Digestion of by the FDA are administered by the IV or SC route. A review of the
ingested collagen is initiated in the stomach via the action of the available information indicates that mAbs are generally adminis-
enzyme pepsin, with subsequent digestion to smaller polypeptides tered to patients in milligram amounts per kilogram of body weight
and ultimately amino acids occurring due to other enzymatic ac- (i.e., mg/kg body weight dose levels), supportive of the targeted
tivity (e.g., trypsin, peptidases, and others) (Hall, 2011b). nature of these therapeutics. Despite their targeted nature, toxic-
No drug products that contain collagen as an active ingredient ities due to mAbs can occur as a result, for example, of their
were identified in the Drugs@FDA database (search conducted April recognized ability to cross the placental barrier and potentially
23, 2015). A search of the U.S. Food and Drug Administration (FDA) cause fetal toxicity. Therefore, although relatively high (milligram)
inactive ingredient database (http://www.accessdata.fda.gov/ quantities are administered therapeutically, an evaluation of all of
scripts/cder/iig/index.cfm; search conducted April 23, 2015) iden- the pharmacology and toxicology data for a given mAb would be
tified three approved drug products in which collagen is present as required to fully appreciate its target effects and potential off-target
an inactive ingredient; these approved drug products include a effects, all of which would impact the derivation of an ADE value.
topical gel, a topical lotion, and a shampoo. The maximum reported One exception to the relatively high doses of mAbs that are typically
potency of collagen in these products is 1%. administered is blinatumomab (Blincyto™; Amgen, Inc.), which is a
Hydrolyzed collagen (also referred to as collagen hydrolysate, newly-developed bi-specific T cell engager (BiTE®; Amgen, Inc.)
gelatin, gelatine, gelatine hydrolysate, and hydrolyzed gelatine) antibody construct that simultaneously targets CD19 on cancer
refers to collagen that has been hydrolyzed from its triple helix cells and CD3 on T cells and is administered in low microgram
conformation into its monomeric components. Safety data for hy- quantities per day. Other emerging technologies are likely to lead to
drolyzed collagen are available, although they relate mainly to the the development of additional novel mAb-based therapeutics that,
oral and dermal routes of administration (CIR, 1985). The oral LD50 like Blincyto™, are more potent than traditional mAbs and are
for hydrolyzed collagen in rats was reported to be >10,000 mg/kg likely to be administered in much lower amounts. The metabolism
body weight while subchronic dermal studies with two cosmetic of mAbs and other biotechnology-derived products is well-known
formulations containing 2% hydrolyzed collagen were negative for and involves degradation to small peptides and individual amino
systemic toxicity (likely due to lack of absorption). Hydrolyzed acids (ICH, 2011).
collagen was found to be non-sensitizing in dermal studies in
guinea pigs (CIR, 1985). More recent evidence from a study in mice 4. Discussion
suggests the potential for oral absorption of small amounts of hy-
drolyzed collagen (Oesser et al., 1999), while food-derived collagen The information on representative substances that is summa-
peptides have been identified in human blood after ingestion of rized above in Section 3 supports the categorization of biologics
gelatin hydrolysates (Iwai et al., 2005). A recent health claims into the four proposed potency bands as suggested by Carver
petition to support the use of collagen hydrolysate in the mainte- (2013). This is outlined in Table 3.
nance of joint health in active individuals was denied by the Eu- A summary of the identified information for the representative
ropean Food Safety Authority on the basis that a cause-and effect substances in each band is as follows.
relationship had not been established between the consumption of Band A: Only low nanogram quantities of these toxins are
collagen hydrolysate and maintenance of joint health (EFSA, 2011). required to exert lethal effects. As such, based on information
In summary, collagen is an endogenous substance that is the identified for the representative substances, this band is considered
most abundant protein in the human body. to represent a very high hazard for cross-contamination.
Band B: Repeated administration of microgram quantities of
3.4.3. Monoclonal antibodies crotoxin (285 mg/day) and TNF-a (270 mg/day) have been demon-
Therapeutic mAbs have been in clinical use for over 20 years strated to be tolerated in humans. However, there are inherent risks
(Hansel et al., 2010; Reichert, 2012). As outlined by Hansel et al. given the foreign nature and potential lethality (crotoxin) or the
(2010), mAbs have high target specificities which facilitate precise recognized pro-inflammatory nature (TNF-a) of these substances.
action, and long half-lives which permit infrequent dosing. More- As such, based on information identified for the representative
over, advances in molecular engineering have enabled fine-tuning substances, this band is considered to represent a high hazard for
of mAb structure for specific therapeutic actions and to minimize cross-contamination.
J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606 601

Table 2
Therapeutic mAbs approved by the U.S. Food and Drug Administration.a

International non-proprietary name First U.S. Indication Dose route Dosing posology
(trade name) approvalb

Rituximab (Rituxan®; MabThera) 1997 Non-Hodgkin's lymphoma IV 375 mg/m2 (~10 mg/kg bw) once
(Biogen Idec Inc.) weekly for 4 or 8 doses.
Chronic lymphocytic leukemia IV 375 mg/m2 (~10 mg/kg bw) in first cycle
and 500 mg/m2 (~13.5 mg/kg bw) in
Cycles 2 to 6.
Rheumatoid arthritis IV 1000 mg on day 0 and 14, repeated
every 24 weeks.
Granulomatosis with Polyangiitis and IV 375 mg/m2 (~10 mg/kg bw) once
Microscopic Polyangiitis weekly for 4 doses.
Daclizumab (Zenapax®) (Hoffmann-La 1997 Prophylaxis of acute organ rejection in IV 1 mg/kg bw every 2 weeks for 5 total
Roche Inc.) renal transplantation doses.
Basiliximab (Simulect®) (Novartis 1998 Prophylaxis of acute organ rejection in IV 20 mg prior to surgery and again 4 days
Pharmaceuticals Corporation) renal transplantation after surgery.
Palivizumab (Synagis®) (MedImmune, 1998 Prevention of RSV disease in children IM 15 mg/kg bw every month during RSV
LLC) season.
Infliximab (Remicade®) (Janssen 1998 Crohn's disease IV 5 mg/kg bw at 0, 2, and 6 weeks, then
Biotech, Inc.) every 8 weeks.
Pediatric Crohn's disease IV 5 mg/kg bw at 0, 2, and 6 weeks, then
every 8 weeks.
Ulcerative colitis IV 5 mg/kg bw at 0, 2, and 6 weeks, then
every 8 weeks.
Pediatric ulcerative colitis IV 5 mg/kg bw at 0, 2, and 6 weeks, then
every 8 weeks.
Rheumatoid arthritis IV 3 mg/kg bw at 0, 2, and 6 weeks, then
every 8 weeks.
Ankylosing spondylitis IV 5 mg/kg bw at 0, 2, and 6 weeks, then
every 6 weeks.
Psoriatic arthritis IV 5 mg/kg bw at 0, 2, and 6 weeks, then
every 8 weeks.
Plaque psoriasis IV 5 mg/kg bw at 0, 2, and 6 weeks, then
every 8 weeks.
®
Trastuzumab (Herceptin ) (Genentech, 1998 HER2 overexpressing breast cancer IV 4 mg/kg bw initially, then 2 mg/kg bw
Inc.) weekly for 52 weeks.
HER2 overexpressing metastatic gastric IV 8 mg/kg bw initially, then 6 mg/kg bw
or gastroesophageal junction every 3 weeks.
adenocarcinoma
Gemtuzumab ozogamicin (Mylotarg®) 2000 CD33 positive acute myeloid leukemia IV 9 mg/m2 (~0.24 mg/kg bw) every 2
(Wyeth Pharmaceuticals Inc.) weeks for 2 total doses.
Alemtuzumab (Lemtrada™; Campath®) 2001 Relapsing multiple sclerosis IV 12 mg/day for 5 days, then 12 mg/day
(Genzyme Corporation) for 3 days (12 months later).
Adalimumab (Humira®) (AbbVie Inc.) 2002 Rheumatoid arthritis SC 40 mg every 2 weeks.
Juvenile idiopathic arthritis SC 10 to 40 mg every 2 weeks.
Psoriatic arthritis SC 40 mg every 2 weeks.
Ankylosing spondylitis SC 40 mg every 2 weeks.
Adult Crohn's disease SC 160 mg on day 1, 80 mg on Day 15, then
40 mg every other week.
Pediatric Crohn's disease SC 80 mg on day 1, 40 mg on Day 15, then
20 mg every other week.
Ulcerative colitis SC 160 mg on day 1, 80 mg on Day 15, then
40 mg every other week.
Plaque psoriasis SC 80 mg on Day 1, then 40 mg every other
week.
®
Ibritumomab tiuxetan (Zevalin ) 2002 Non-Hodgkin's lymphoma IV 250 mg/m2 (~6.8 mg/kg bw) on Day 1
(Spectrum Pharmaceuticals, Inc.) and Day 7, 8, or 9.
Efalizumab (Raptiva®) (Genentech, Inc.) 2003 Plaque psoriasis SC 0.7 mg/kg bw initial dose, then 1 mg/kg
every week.
Tositumomab-I131 (Bexxar®) 2003 Non-Hodgkin's lymphoma IV 450 mg once weekly for 2 doses.
(GlaxoSmithKline)
Omalizumab (Xolair®) (Novartis AG) 2003 Moderate to severe persistent asthma SC 150 to 375 mg every 2 or 4 weeks.
Chronic idiopathic urticaria SC 150 or 300 mg every 4 weeks.
®
Cetuximab (Erbitux ) (ImClone LLC) 2004 Head and neck cancer IV 400 mg/m2 (~10.8 mg/kg bw) followed
by weekly doses of 250 mg/m2
(~6.8 mg/kg bw).
Colorectal cancer IV 400 mg/m2 (~10.8 mg/kg bw) followed
by weekly doses of 250 mg/m2
(~6.8 mg/kg bw).
Bevacizumab (Avastin®) (Genentech, 2004 Metastatic colorectal cancer IV 5 mg/kg bw every 2 weeks.
Inc.) Non-squamous non-small cell lung IV 15 mg/kg bw every 3 weeks.
cancer
Glioblastoma IV 10 mg/kg bw every 2 weeks.
Metastatic renal cell carcinoma IV 10 mg/kg bw every 2 weeks.
Cervical cancer IV 15 mg/kg bw every 3 weeks.
(continued on next page)
602 J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606

Table 2 (continued )

International non-proprietary name First U.S. Indication Dose route Dosing posology
(trade name) approvalb

Platinum-resistant recurrent epithelial IV 10 mg/kg bw every 2 weeks or 15 mg/


ovarian, fallopian tube or primary kg bw every 3 weeks.
peritoneal cancer
Natalizumab (Tysabri®) (Biogen Idec 2004 Multiple sclerosis IV 300 mg every 4 weeks.
Inc.) Crohn's disease IV 300 mg every 4 weeks.
Ranibizumab (Lucentis®) (Genentech, 2006 Neovascular (wet) age-related macular Intravitreal 0.5 mg every month.
Inc.) degeneration
Macular edema following retinal vein Intravitreal 0.5 mg every month.
occlusion
Diabetic macular edema Intravitreal 0.3 mg every month.
Diabetic retinopathy in patients with Intravitreal 0.3 mg every month.
diabetic macular edema
Panitumumab (Vectibix®) (Amgen Inc.) 2006 Metastatic colorectal cancer IV 6 mg/kg bw every 2 weeks.
Eculizumab (Soliris®) (Alexion 2007 Paroxysmal nocturnal hemoglobinuria IV 600 mg every week for 4 weeks, then
Pharmaceuticals, Inc.) 900 mg for Week 5, then 900 mg every
2 weeks.
Atypical hemolytic uremic syndrome IV 900 mg every week for 4 weeks, then
1200 mg for Week 5, then 1200 mg
every 2 weeks.
Golimumab (Simponi®; Simponi® 2009 Rheumatoid arthritis IV 2 mg/kg bw at Weeks 0 and 4, then
ARIA™) (Janssen Biotech, Inc.) 2 mg/kg bw every 8 weeks.
Rheumatoid arthritis SC 50 mg every month.
Active psoriatic arthritis SC 50 mg every month.
Active ankylosing spondylitis SC 50 mg every month.
Ulcerative colitis SC 200 mg initial dose, 100 mg at week 2,
then 100 mg every 4 weeks.
Canakinumab (Ilaris®) (Novartis 2009 Cryopyrin-associated periodic SC 150 mg every 8 weeks.
Pharmaceuticals Corporation) syndrome
Systemic juvenile idiopathic arthritis SC 4 mg/kg bw every 4 weeks.
Ofatumumab (Arzerra®) 2009 Chronic lymphocytic leukemia IV 300 mg first dose, followed by 2000 mg
(GlaxoSmithKline) weekly for 7 doses, then 2000 mg every
4 weeks for 4 doses.
Ustekinumab (Stelara®) (Janssen 2009 Moderate to severe plaque psoriasis SC 45 mg initial, 45 mg at Week 4, then
Biotech, Inc.) 45 mg every 12 weeks.
Active plaque arthritis SC 45 mg initial, 45 mg at Week 4, then
45 mg every 12 weeks.
Tocilizumab (Actemra®) (Genentech, 2010 Rheumatoid Arthritis in adults IV 4 mg/kg bw every 4 weeks.
Inc.) Rheumatoid Arthritis in adults SC 162 mg bw every 2 weeks.
Polyarticular Juvenile Idiopathic IV 8 mg/kg bw every 4 weeks.
Arthritis
Systemic Juvenile Idiopathic Arthritis IV 8 mg/kg bw every 2 weeks.
Denosumab (Prolia®; Xgeva®) (Amgen, 2010 Postmenopausal women with SC 60 mg once every 6 months.
Inc.) osteoporosis at high risk for fracture
Increase bone mass in men with SC 60 mg once every 6 months.
osteoporosis
Bone loss in men receiving androgen SC 60 mg once every 6 months.
deprivation therapy for prostate cancer
Bone loss in women receiving adjuvant SC 60 mg once every 6 months.
aromatase therapy for breast cancer
Belimumab (Benlysta®) 2011 Systemic lupus erythmatosus IV 10 mg/kg bw every 2 weeks for 3 doses,
(GlaxoSmithKline) then once every 4 weeks.
Ipilimumab (Yervoy®) (Briston-Myers 2011 Unresectable or metastatic melanoma IV 3 mg/kg bw every 3 weeks for total of 4
Squibb Company) doses.
Brentuximab vedotin (Adcetris®) 2011 Hodgkin lymphoma IV 1.8 mg/kg bw every 3 weeks.
(Seattle Genetics) Anaplastic large cell lymphoma IV 1.8 mg/kg bw every 3 weeks.
Raxibacumab (RAXIBACUMAB) 2012 Inhalational anthrax IV 40 mg/kg bw single dose.
(GlaxoSmithKline)
Pertuzumab (Perjeta®) (Genentech, 2012 HER2-positive breast cancer IV 840 mg initial dose, then 420 mg every
Inc.) 3 weeks.
Obinutuzumab (Gazyva™) (Genentech, 2013 Chronic lymphocytic leukemia IV Cycle 1: 100 mg on Day 1, 900 mg on
Inc.) Day 2, 1000 mg on Day 8 and 15; Cycles
2 to 6: 1000 mg on Day 1.
Ado-Trastuzumab Emtansine 2013 HER2-positive metastatic breast cancer IV 3.6 mg every 3 weeks.
(Kadcyla™) (Genentech, Inc.)
Blinatumomab (Blincyto™) (Amgen, 2014 Philadelphia chromosome-negative IV Cycle 1: 9 mg/day on Days 1 to 7, 28 mg/
Inc.) relapsed or refractory B-cell precursor day on Days 8 to 28; Subsequent cycles:
acute lymphoblastic leukemia 28 mg/day for 28 days.
Nivolumab (Opdivo®) (Bristol-Myers 2014 Unresectable or metastatic melanoma IV 3 mg/kg bw every 2 weeks.
Squibb Company) Metastatic squamous non-small cell IV 3 mg/kg bw every 2 weeks.
lung cancer
Pembrolizumab (Keytruda®) (Merck & 2014 Unresectable or metastatic melanoma IV 2 mg/kg bw every 3 weeks.
Co., Inc.)
J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606 603

Table 2 (continued )

International non-proprietary name First U.S. Indication Dose route Dosing posology
(trade name) approvalb

Ramucirumab (Cyramza®) (Eli Lilly and 2014 Advanced or metastatic gastric or IV 8 mg/kg bw every 2 weeks.
Company) gastro-esophageal junction
adenocarcinoma
Metastatic non-small cell lung cancer IV 10 mg/kg bw on Day 1 of a 21-day cycle.
(in combination with docetaxel)
Siltuximab (Sylvant™) (Janssen Biotech, 2014 Multicentric Castleman's disease IV 11 mg/kg bw every 3 weeks.
Inc.)
Vedolizumab (Entyvio™) (Takeda 2014 Adult ulcerative colitis IV 300 mg at 0, 2, and 6 weeks and every 8
Pharmaceuticals America, Inc.) weeks thereafter.
Adult Crohn's disease IV 300 mg at 0, 2, and 6 weeks and every 8
weeks thereafter.
Secukinumab (Cosentyx™) (Novartis 2015 Moderate to severe plaque psoriasis SC 300 mg at 0, 1, 2, 3, and 4 weeks and
Pharmaceuticals Corporation) every 4 weeks thereafter.
Dinutuximab (Unituxin™) (United 2015 Pediatric high-risk neuroblastoma IV 17.5 mg/m2/day for 4 consecutive days
Therapeutics Corp.) (over 10 to 20 h/day) for a maximum of
5 cycles (cycle days and durations are
variable).

bw ¼ body weight; HER2 ¼ human epidermal growth factor receptor 2; IM ¼ intramuscular; IV ¼ intravenous; RSV ¼ respiratory syncytial virus; SC ¼ subcutaneous.
a
Only therapeutic monoclonal antibodies and monoclonal antibodyedrug conjugates are included. Not included are diagnostic products or products that are based on
fragment antigen-binding (Fab) regions of antibodies [e.g., abciximab (ReoPro®) and certolizumab (Cimzia®)].
b
Listed chronologically.

Band C: These cytokines and growth factors are endogenous the proposed potency bands were identified by Carver (2013), but
substances. Recombinant human versions of these substances are the potency/toxicity data that have been identified in the current
administered to patients in low (interferons), intermediate (growth assessment would seem to support the overall approach, at least in
factors), and high (interleukins) microgram quantities for the general terms. The results of this evaluation indicate that the ulti-
treatment of various disease conditions, sometimes on a chronic mate designation of a given substance into any of the proposed
basis. As such, based on information identified for the representa- potency bands will require a combination of quantitative and
tive substances, this band is considered to represent a medium qualitative data and information related to its source (endogenous
hazard for cross-contamination. vs. foreign), potency, toxicity, allergenic potential, and other factors.
Band D: These substances are endogenous and are present or It should be noted that the calculated “maximum daily expo-
produced in the human body in milligram quantities per day (in- sure” values that are presented in Table 3 represent established
sulin, collagen), or are protein therapeutics that are administered in dose/exposure levels for the representative substances (e.g., known
milligram quantities but are targeted to specific receptors within toxic or therapeutic doses) and are not synonymous with ADE
the body and therefore are associated with minimal off-target ef- values (or PDE values as they are more commonly known). While
fects (mAbs). It is anticipated that calculated ADE values for mAb ADE values for the four proposed potency bands are presented in
products would still be higher than those for substances in other Figs. 2 and 3 of the recent publication by Carver (2013), it is stated
potency bands, even after adjusting for the intermittent dosing in the publication that these ADE values are “indicative and are only
schedules and prolonged half-lives that are typical of therapeutic presented for illustrative purposes”. Indeed, as outlined by Carver
mAb products. As such, based on information identified for the (2013), an ADE value for a given biologic is a value that is consid-
representative substances, this band is considered to represent a ered safe for any and all potential “recipients” of the biologic as an
low hazard for cross-contamination. unintended impurity in another product. As such, in addition to a
It is recognized that there is some overlap for some of the lack of toxicological potential, there shouldn't be any meaningful
substances in Bands B and C with regard to the estimated exposure pharmacological activity at the ADE because what is efficacious (i.e.,
(i.e., daily dose) values that are presented in Table 3. Specifically, the desired) for a biologic in an intended patient may be detrimental in
calculated maximum daily exposure values for crotoxin and TNF-a, someone else who receives a small quantity of the biologic as an
which are representative substances for Band B, are somewhat impurity.
higher than the calculated maximum daily exposure values for the To derive an ADE value for any given substance would require a
interferons (IFNa-2b, IFNb-1a, and IFNg-1b) that are representative complete dataset for that substance (pharmacology data, pharma-
substances in Band C. It should be noted, however, that these in- cokinetics information, full toxicology profile, etc.); this would
terferons are administered to patients once or several times per represent a significant effort which is beyond the scope of the
week on a continual basis for the treatment of chronic diseases (e.g., current assessment. In addition, much of this information is usually
IFNa-2b for the treatment of chronic Hepatitis C), and single or lacking in early development programs.
shorter-term exposures to higher levels likely would be tolerable. Based on the information identified in this proof-of-concept
Conversely, the representative substances for Band B (crotoxin and exercise, it is anticipated that if sufficient information was avail-
TNF-a) have been evaluated only in a limited number of clinical able to calculate ADE values for each of the representative sub-
trials of relatively short duration, and it is possible (and perhaps stances from each potency band, the ADE values would align in a
likely) that longer exposure periods would only be tolerable if the similar manner as the maximum daily exposure values that are
exposure level were to be decreased. Given this information, it is presented above in Table 3. In other words, it is expected that the
considered appropriate to classify crotoxin and TNF-a as Band B ADE values for Band A substances would be lowest (most potent/
substances and the interferons as Band C substances despite the toxic substances) and the ADE values for Band D substances would
apparent similarity in numerical exposure values (as presented in be highest (least potent/toxic substances). This would be of prac-
Table 3). tical benefit since maximum daily exposure values would be easier
It is not known how the representative substances for each of to estimate for early development programs and more amenable
604 J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606

Table 3
Summary of information to support the 4 proposed potency bands.

Potency Representative Noteworthy information on potency/ Effect level/dose level Maximum daily exposure as a toxin
band substance toxicity (Band A) or as an endogenous substance
or therapeutic agent (Bands B, C, and D)
for a 50 kg patienta

A Botulinum toxin Potent bacterial toxin. Low ng/kg body weight range Low nanogram amount
(estimated IV or IP lethal dose).
Diphtheria toxin Potent bacterial toxin. Low ng/kg body weight range Low nanogram amount
(estimated IV lethal dose).
B Crotoxin Snake venom toxin. Low mg/kg body weight range Low microgram amount
Evaluated in a Phase 1 clinical trial in (estimated IV lethal dose).
cancer patients. Up to 5.7 mg/kg body weight/day for 30 285 mg
days evaluated in patients (IM
injection).
TNF-a Endogenous pyrogen with multiple 4 to 5.4 mg/kg body weight range 270 mg
physiological functions and a central generally considered to be the MTD on
role in several autoimmune diseases. various dosing schedules (IV infusion).
Evaluated in multiple Phase 1 and 2
clinical trials in cancer patients with
expected dose-limiting toxicities
(hypotension, fever, etc.).
C IFNa-2b Endogenous cytokine with anti-viral 1.5 mg/kg body weight/week (SC 75 mg
properties. injection).
Recombinant version is used to treat
chronic Hepatitis C.
IFNb-1a Endogenous cytokine with Up to 44 mg three times per week (SC 44 mg
immunomodulatory, anti-viral, and injection).
anti-proliferative properties.
Recombinant version is used to treat
relapsing forms of multiple sclerosis.
IFNg-1b Endogenous cytokine that activates 1.35 mg/kg body weight three times per 67.5 mg
macrophages and has anti-viral, week (SC injection).
immunoregulatory, and anti-tumor
properties.
Recombinant version is used to reduce
frequency and severity of serious
infections associated with Chronic
Granulomatous Disease and for
delaying time to disease progression in
patients with severe, malignant
osteoporosis.
IL-2 Endogenous cytokine with numerous 37 mg/kg body weight every 8 h by 15- 5550 mg
immunoregulatory properties. min IV infusion for a total of 14 doses;
Recombinant version is used to treat repeated after 9 days of rest for a total of
adults with renal cell carcinoma or 28 doses per course, as tolerated.
metastatic melanoma.
IL-11 Endogenous cytokine that stimulates 50 mg/kg body weight/day for a 2500 mg
proliferation of hematopoietic stem maximum of 21 days per treatment
cells and megakaryocyte progenitor course (SC injection).
cells and induces megakaryocyte
maturation resulting in increased
platelet production.
Recombinant version is used to prevent
severe thrombocytopenia and reduce
the need for platelet transfusions
following myelosuppressive
chemotherapy in adult patients with
non-myeloid malignancies who are at
high risk of severe thrombocytopenia.
G-CSF Endogenous growth factor that Dose level depends on indication. 250 mg
stimulates proliferation, differentiation For patients with cyclic or idiopathic
commitment, and some end-cell neutropenia, the recommended dose is
functional activation of neutrophils. 5 mg/kg body weight/day (SC injection)
Recombinant version is used to
stimulate neutrophil production in
various indications.
GM-CSF Endogenous growth factor that 6.75 mg/kg body weight/day (IV 337.5 mg
promotes the number and function of infusion; duration varies depending on
white blood cells, including the indication).
neutrophils, monocytes/macrophages,
and dendritic cells.
Recombinant version is used to
stimulate white blood cell production in
various indications.
D Insulin Endogenous peptide hormone that 360 to 1800 mg/day in children (10 kg) 1800 mg (normal background)
regulates blood glucose levels. and adults (50 kg), respectively 1900 mg (therapeutic in diabetics)
J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606 605

Table 3 (continued )

Potency Representative Noteworthy information on potency/ Effect level/dose level Maximum daily exposure as a toxin
band substance toxicity (Band A) or as an endogenous substance
or therapeutic agent (Bands B, C, and D)
for a 50 kg patienta

Several human insulin drug products (“background” rates of secretion from


are available. the pancreas).
190 to 380 mg/day (10 kg child) and
950e1900 mg/day (50 kg adult) (total
daily insulin requirement for diabetics).
Collagen Endogenous compound produced N/A N/A
primarily by fibroblasts.
It is the most abundant protein in the
human body.
Information was not identified on the
presence of collagen in blood, although
markers of collagen degradation can be
measured in blood and urine.
mAbs Targeted protein therapeutics used to Dose levels in the mg/kg body weight Several milligrams
treat various diseases. range are typically utilized on various
dosing schedules (usually SC or IV
route).

G-CSF ¼ granulocyte colony stimulating factor; GM-CSF ¼ granulocyte-macrophage colony stimulating factor; IFN ¼ interferon; IL ¼ interleukin; IM ¼ intramuscular;
IP ¼ intraperitoneal; IV ¼ intravenous; mAbs ¼ monoclonal antibodies; MTD ¼ maximum tolerated dose; SC ¼ subcutaneous; TNF ¼ tumor necrosis factor.
a
Refers to the dose level on any given day and is not intended to imply daily dosing.

for use in their risk assessments. Moreover, this would be funda- Carver on his work in this area.
mentally similar to the banding scheme and corresponding ADE
values that were derived by Dolan et al. (2005) for small molecule Transparency document
drug substances, although it is presently unknown whether ADE
values for different categories of biologics and small molecules Transparency document related to this article can be found
might overlap. online at http://dx.doi.org/10.1016/j.yrtph.2015.09.003.

5. Conclusions
References
Based on an evaluation of information that was identified for the Barksdale, L., 1970. Corynebacterium diphtheriae and its relatives. Bacteriol. Rev. 34,
representative substances, it is considered that the banding scheme 378e422.
proposed by Carver (2013) is a feasible approach to provide an Bercu, J.P., Sharnez, R., Dolan, D.G., 2013. Advancing toxicology in RiskMAPP: setting
ADEs based on the subsequent drug substance. Regul. Toxicol. Pharmacol. 65,
initial evaluation of risks associated with inadvertent exposure to 157e161.
biologics as potential carryover impurities in other products. Sub- Cardoso, D.F., Lopes-Ferreira, M., Faquim-Mauro, E.L., Macedo, M.S., Farsky, S.H.,
stances in Bands A, B, C, and D are considered to represent very 2001. Role of crotoxin, a phospholipase A2 isolated from Crotalus durissus
terrificus snake venom, on inflammatory and immune reactions. Mediat.
high, high, medium, and low risk, respectively, with regard to po- Inflamm. 10, 125e133.
tential cross-contamination in manufacturing facilities. The ulti- Carver, M., Nov. 2013. Applying a Health- and Science-based Risk Assessment for
mate designation of a given substance into any of the proposed Multi-product Biologics Manufacturing. (White Paper). Fujifilm Diosynth Bio-
technologies UK Limited. Available at: http://www.outsourcing-pharma.com/
potency bands will require consideration of a combination of smartlead/view/846676/4/Applying-a-Health-and-Science-based-Risk-
quantitative and qualitative data and information related to a Assessment-for-Multi-Product-Biologics-manufacturing.
substance's source (endogenous vs. foreign), potency, toxicity, Chu, W.M., 2013. Tumor necrosis factor. Cancer Lett. 328, 222e225.
CIR, 1985. Final report on the safety assessment of hydrolyzed collagen (cosmetic
allergenic potential, and other factors.
ingredient review). Int. J. Toxicol. 4, 199e221.
The approach outlined by Carver (2013) can be utilized to help Cura, J.E., Blanzaco, D.P., Brisson, C., Cura, M.A., Cabrol, R., Larrateguy, L., et al., 2002.
establish safe exposure limits that in turn can be used to assist with Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA2, NSC-624244)
in patients with advanced cancer. Clin. Cancer Res. 8, 1033e1041.
cleaning validation efforts, similar to what has been proposed for
Di Lullo, G.A., Sweeney, S.M., Korkko, J., Ala-Kokko, L., San Antonio, J.D., 2002.
APIs (ISPE, 2010; Walsh, 2011a,b,c; Bercu et al., 2013; Sargent et al., Mapping the ligand-binding sites and disease-associated mutations on the
2013). It should be noted, however, that the purpose of this eval- most abundant protein in the human, type I collagen. J. Biol. Chem. 277,
uation was not to derive ADE values for any of the representative 4223e4231.
Dolan, D.G., Naumann, B.D., Sargent, E.V., Maier, A., Dourson, M., 2005. Application
substances or to establish absolute upper and lower exposure limits of the threshold of toxicological concern concept to pharmaceutical
for each of the proposed bands. Rather, the purpose was to deter- manufacturing operations. Regul. Toxicol. Pharmacol. 43, 1e9.
mine whether information was available to support the proposed EFSA, 2011. EFSA Panel on Dietetic Products, Nutrition and Allergies; Scientific
Opinion on the substantiation of a health claim related to collagen hydrolysate
banding scheme from a scientific perspective, based on a limited and maintenance of joints pursuant to Article 13(5) of Regulation (EC) No 1924/
assessment of representative substances from each band. Given the 2006 (question no EFSA-Q-2011-00201, adopted on 30 June 2011 by European
vast number of substances that may fall into each of the proposed Food Safety Authority). EFSA J. 9 (2291), 11. http://dx.doi.org/10.2903/
j.efsa.2011.2291. Available at: http://www.efsa.europa.eu/en/efsajournal/pub/
bands, additional detailed work beyond this initial assessment 2291.htm (last accessed 08.09.15.).
would be required to allow for derivation of ADE values for the EMA, Nov. 20, 2014. Guideline on Setting Health Based Exposure Limits for Use in
representative substances and, if possible, delineation of absolute Risk Identification in the Manufacture of Different Medicinal Products in Shared
Facilities. (EMA/CHMP/CVMP/SWP/169430/2012). European Medicines Agency
upper and lower exposure limits for each of the proposed bands.
(EMA), Committee for Medicinal Products for Human Use (CHMP) & Committee
for Medicinal Products for Veterinary Use (CVMP), London, UK. Available at:
Acknowledgments http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_
guideline/2014/11/WC500177735.pdf (last accessed 08.09.15.).
Gill, D.M., 1982. Bacterial toxins: a table of lethal amounts. Microbiol. Rev. 46,
The authors appreciate discussion with and input from Dr. M. 86e94.
606 J.W. Card et al. / Regulatory Toxicology and Pharmacology 73 (2015) 595e606

Hadfield, T.L., McEvoy, P., Polotsky, Y., Tzinserling, V.A., Yakovlev, A.A., 2000. The iss), D959eD965. Available at: http://www.plasmaproteomedatabase.org/
pathology of diphtheria. J. Infect. Dis. 181 (Suppl. 1), S116eS120. index.html (last accessed 08.09.15.).
Hall, J.E., 2011a. Insulin, glucagon, and diabetes mellitus. In: Guyton and Hall NIBSC, 2010. WHO International Standard Insulin, Human NIBSC Code: 83/500 In-
Textbook of Medical Physiology, 12th ed. W.B. Saunders Company, Elsevier, structions for Use. (Version 4.0, Dated 23/09/2010). U.K. Medicines and
Philadelphia (PA), pp. 939e954. Healthcare Products Regulatory Agency, National Institute for Biological Stan-
Hall, J.E., 2011b. Digestion and absorption in the gastrointestinal tract. In: Guyton dards and Control (NIBSC)/WHO International Laboratory for Biological Stan-
and Hall Textbook of Medical Physiology, 12th ed. W.B. Saunders Company, dards, UK Official Medicines Control Laboratory, Potters Bar, Hertfordshire, UK.
Elsevier, Philadelphia (PA), pp. 789e798. Available at: http://www.nibsc.org/documents/ifu/83-500.pdf (last accessed
Hansel, T.T., Kropshofer, H., Singer, T., Mitchell, J.A., George, A.J., 2010. The safety and 08.09.15.).
side effects of monoclonal antibodies. Nat. Rev. Drug Discov. 9, 325e338. Oesser, S., Adam, M., Babel, W., Seifert, J., 1999. Oral administration of (14) C labeled
ICH, 2011. Preclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals: gelatin hydrolysate leads to an accumulation of radioactivity in cartilage of mice
S6(R1): Current Step 4 version Addendum dated 12 June 2011 incorporated at (C57/BL). J. Nutr. 129, 1891e1895.
the end of June 2011. (ICH Harmonised Tripartite Guideline, Parent Guideline Reichert, J.M., 2012. Marketed therapeutic antibodies compendium. MAbs 4,
dated 16 July 1997). In: Geneva, Switz.: International Conference on Harmo- 413e415.
nisation of Technical Requirements for Registration of Pharmaceuticals for Rennie, M.J., 1999. Teasing out the truth about collagen. J. Physiol. 521, 1.
Human Use (ICH). Available at: http://www.ich.org/fileadmin/Public_Web_Site/ Roberts, N.J., Zhou, S., Diaz Jr., L.A., Holdhoff, M., 2011. Systemic use of tumor ne-
ICH_Products/Guidelines/Safety/S6_R1/Step4/S6_R1_Guideline.pdf (last crosis factor alpha as an anticancer agent. Oncotarget 2, 551e739.
accessed 08.09.15.). Sargent, E.V., Faria, E., Pfister, T., Sussman, R.G., 2013. Guidance on the establish-
ISPE, 2010. Risk-based Manufacture of Pharmaceutical Products: a Guide to Man- ment of acceptable daily exposure limits (ADE) to support risk-based manu-
aging Risks Associated with Cross-contamination. In: Baseline Pharmaceutical facture of pharmaceutical products. Regul. Toxicol. Pharmacol. 65, 242e250.
Engineering Guide, vol. 7. ISPE Headquarters, Tampa (FL). Shapira, A., Benhar, I., 2010. Toxin-based therapeutic approaches. Toxins 2,
Iwai, K., Hasegawa, T., Taguchi, Y., Morimatsu, F., Sato, K., Nakamura, Y., et al., 2005. 2519e2583.
Identification of food-derived collagen peptides in human blood after oral Smith, K., Rennie, M.J., 2007. New approaches and recent results concerning
ingestion of gelatin hydrolysates. J. Agric. Food Chem. 53, 6531e6536. human-tissue collagen synthesis. Curr. Opin. Clin. Nutr. Metab. Care 10,
Kotsonis, F.N., Burdock, G.A., 2008. Food toxicology (chapter 30). In: Klaassen, C.D. 582e590.
(Ed.), Casarett and Doull's Toxicology: the Basic Science of Poisons, seventh ed. Sobel, J., 2005. Botulism. Food Saf. 41, 1167e1173.
McGraw-Hill, Medical Publishing Division, New York (NY)/Toronto (ON), Walsh, A., 2011a. Cleaning validation for the 21st century: acceptance limits for
pp. 1191e1236. active pharmaceutical ingredients (APIs): part I. Pharm. Eng. 31 (5), 74e83.
Mott, A., Henry, B., Wyman, E., Bellorado, K., Blumel, M., Parks, M., et al., 2013. Walsh, A., 2011b. Cleaning validation for the 21st century: acceptance limits for
Methodology for assessing product inactivation during cleaning part II: setting active pharmaceutical ingredients (APIs): part II. Pharm. Eng. 31 (4), 44e49.
acceptance limits of biopharmaceutical product carryover for equipment Walsh, A., 2011c. Cleaning validation for the 21st century: overview of new ISPE
cleaning. J. Valid. Technol. 4. cleaning guide. Pharm. Eng. 31 (6), 1e7.
Myers, A.K., Robey, J.W., Price, R.M., 1990. Relationships between tumour necrosis Watkins, J.B., 2008. Properties and toxicities of animal venoms (chapter 26). In:
factor, eicosanoids and platelet-activating factor as mediators of endotoxin- Klaassen, C.D. (Ed.), Casarett and Doull's Toxicology: the Basic Science of Poi-
induced shock in mice. Br. J. Pharmacol. 99, 499e502. sons, seventh ed. McGraw-Hill, Medical Publishing Division, New York (NY)/
Nanjappa, V., Thomas, J.K., Marimuthu, A., Muthusamy, B., Radhakrishnan, A., Toronto (ON), pp. 1083e1102.
Sharma, R., et al., 2014. Plasma proteome database as a resource for proteomics Yamaizumi, M., Mekada, E., Uchida, T., Okada, Y., 1978. One molecule of diphtheria
research: 2014 update. Tumor necrosis factor Nucleic Acids Res. 42 (database toxin fragment A introduced into a cell can kill the cell. Cell 15, 245e250.

You might also like