Skip to main content
Oncoimmunology logoLink to Oncoimmunology
. 2013 Oct 17;2(10):e26333. doi: 10.4161/onci.26333

Adverse events to monoclonal antibodies used for cancer therapy

Focus on hypersensitivity responses

Brian A Baldo 1,2,†,*
PMCID: PMC3827071  PMID: 24251081

Abstract

Fifteen monoclonal antibodies (mAbs) are currently registered and approved for the treatment of a range of different cancers. These mAbs are specific for a limited number of targets (9 in all). Four of these molecules are indeed directed against the B-lymphocyte antigen CD20; 3 against human epidermal growth factor receptor 2 (HER2 or ErbB2), 2 against the epidermal growth factor receptor (EGFR), and 1 each against epithelial cell adhesion molecule (EpCAM), CD30, CD52, vascular endothelial growth factor (VEGF), tumor necrosis factor (ligand) superfamily, member 11 (TNFSF11, best known as RANKL), and cytotoxic T lymphocyte-associated protein 4 (CTLA4). Collectively, the mAbs provoke a wide variety of systemic and cutaneous adverse events including the full range of true hypersensitivities: Type I immediate reactions (anaphylaxis, urticaria); Type II reactions (immune thrombocytopenia, neutopenia, hemolytic anemia); Type III responses (vasculitis, serum sickness; some pulmonary adverse events); and Type IV delayed mucocutaneous reactions as well as infusion reactions/cytokine release syndrome (IRs/CRS), tumor lysis syndrome (TLS), progressive multifocal leukoencephalopathy (PML) and cardiac events. Although the term “hypersensitivity” is widely used, no common definition has been adopted within and between disciplines and the requirement of an immunological basis for a true hypersensitivity reaction is sometimes overlooked. Consequently, some drug-induced adverse events are sometimes incorrectly described as “hypersensitivities” while others that should be described are not.

Keywords: adverse effects, cancer, cardiac events, hemolytic anemia, hypersensitivity, infusion reactions, lung disease, mAbs, monoclonal antibodies, neutropenia, serum sickness, thrombocytopenia, vasculitis

Adverse and Hypersensitivity Reactions to Drugs

Over the years a number of different definitions have been advanced to describe an adverse drug reaction. The most quoted one is that issued by the World Health Organization in 1972, that is, “a response to a drug that is noxious and unintended and occurs at doses normally used in man for the prophylaxis, diagnosis or therapy of disease, or for modification of physiological function.”1 Disagreements with this definition, for example, with use of the words “noxious” and “drug,” have led to a number of other descriptions. That advanced by Edwards and Aronson2—“an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product, which predicts hazard from future administration and warrants prevention or specific treatment, or alteration of the dosage regimen, or withdrawal of the product”—is often referred to, but for many the simplicity of the United States Food and Drug Administration (FDA) definition—“any undesirable experience associated with the use of a medical product in a patient”—is adequate. The FDA states that any serious event induced by a medical product should be reported to the organization and defines “serious” as death, life-threatening, hospitalization, disability or permanent damage, congenital anomaly/birth defect, required intervention to prevent permanent impairment or damage (devices), and other serious important medical events (for example, allergic bronchospasm, serious blood dyscrasias, or seizures or convulsions that do not result in hospitalization).3

Adverse drug reactions (ADRs) have been classified into seven different categories, designated A to G. These categories are: A, Augmented pharmacologic effects; B, Bizarre; C, Chronic (or continuous) effects; D, Delayed effects; E, End-of-treatment (or withdrawal) effects; F, Failure of therapy; and G, Genetic reactions.2,4 Type A reactions, distinguished primarily on the basis of dose-related reactions, are predictable, can be anticipated from the drug's pharmacological activity, resolve when the dose is reduced or withdrawn and account for ~80% of ADRs. For our purposes, type B reactions are the main concern. Type B reactions are non-dose-related, unpredictable, generally unrelated to the drug's pharmacological activity, and usually resolve when treatment is terminated. These reactions are divided into true allergic responses (immune-mediated) and non-allergic (non-immune) sensitivities (Table 1). The former category is made up of the 4 true hypersensitivity states, as defined by Gell and Coombs: Type I, IgE antibody-mediated reactions; Type II, cytotoxic reactions; Type III, immune-complex-mediated hypersensitivities; and Type IV, delayed, cell-mediated responses.5 Conversely, pseudoallergy, idiosyncratic reactions and Type B intolerances make up Type B non-immune, non-allergic drug sensitivities (Table 1). The term “hypersensitivity” is often misused. Here, it is taken to mean adverse signs and symptoms that are initiated by an antigenic stimulus tolerated by a “normal” person and that have an immune basis or component. Even in the absence of evidence of an immune mechanism, a few drugs show apparent features of a hypersensitivity response. Some reactions to contrast media6 and non-steroidal anti-inflammatory drugs (NSAIDs)7 fall into this group, in which mechanisms such as the activation of bradykinin and the drug-induced redirection of the arachidonic acid metabolism from the cyclooxygenase to the lipoxygenase pathway may be involved. Such reactions are described here as non-immune “sensitivities” or “intolerances.”8

Table 1. Classification of Type Ba adverse reactions to therapeutic agents.

Hypersensitivity
(Immune; Allergic)
Non-Allergic Sensitivities
(Non-immune)
Hypersensitivity
Typeb
Mediated by
Examples
Subtypes
Examples
Type Ic
IgE
antibodies
Penicillins; cephalosporins; neuromuscular blockers; mAbs; pyrazolones; proton pump inhibitors; L-asparaginase
Pseudo-
Allergyd
Most reactions to NSAIDs;e
Direct mast cell degranulation (neuromuscular blockers; opioids; contrast media; vancomycin)
Type IIf
IgG/IgM cytotoxic reactions
Penicillins; cephalosporins; quinine; quinidine; sulfonamides; thiouracil; oxaliplatin; mAbs; gold salts; NSAIDs; procainamide; ticlopidine
Idiosyncratic Reactionsg
Halothane hepatitis; malignant hypothermia; drug induced hemolytic anemia in Glu-6-PO4 dehydrogenase-deficient individuals (anti-malarials, sulfonamides, dapsone)
Type IIIh
IgG/IgM immune complexes
Penicillins; cephalosporins; sulfonamides; allopurinol; NSAIDs; carbamazepine; mAbs; tamoxifen; oxaliplatin
Intolerances ? some reactions to contrast media and NSAIDsi
Tinnitus induced by small doses of aspirin
Type IVj,k T cells NSAIDs; penicillins; local anesthetics; hydroxychloroquine; anti-convulsants (eg carbamazepine); dapsone; mAbs; tamoxifen

Note that mAbs can provoke all 4 types of hypersensitivity reactions. aSo-called ‘Bizarre’ reactions that are uncommon unpredictable, rarely dose dependent and unrelated to agent’s pharmacologic action. Relative to other categories of adverse reactions, these reactions show high mortality. bAccording to the definition and classification of Gell and Coombs (see ref.5). cManifest as anaphylaxis, bronchospasm, cardiovascular collapse, urticaria, angioedema. dSome reactions closely resemble true Type I reactions and are termed 'anaphylactoid'. eNSAIDs, non-steroidal anti-inflammatory drugs. fE.g. drug-induced hemolytic anemia, immune thrombocytopenia, immune form of agranulocytosis. gMay be unrelated or related to dose. Uncommon, unpredictable, unrelated to drug’s pharmacologic action. hE.g. serum sickness-like reactions, drug-induced vasculitis. iSome reactions to NSAIDs and contrast media are clearly not hypersensitivity responses or pseudoallergic or idiosyncratic in nature. jE.g. allergic contact dermatitis, psoriasis, maculopapular exanthema, acute generalized exanthematous pustulosis, drug reaction with eosinophilia and systemic symptoms, erythema multiforme, fixed drug eruption, Stevens–Johnson syndrome, toxic epidermal necrolysis. kAlso known as delayed reactions.

Even though the term “hypersensitivity” is widely used including, for example, in the immunology, allergy, oncology and pharmacology literatures as well as in clinical trial reports, company drug descriptions and drug package inserts, no common definition has been adopted. The word is sometimes mistakenly used to cover reactions that clearly have no immune basis and which are otherwise not easy to classify, or where the mechanism remains to be determined.9 On the other hand, since the mechanisms accounting for many ADRs are not known or not investigated, and appropriate tests are either not at hand or even developed, some true hypersensitivities remain unrecognized and therefore incorrectly classified. It is not uncommon therefore to see adverse reactions incorrectly described as “hypersensitivities”10 as well as the use of alternative terms where that of “hypersensitivity” would be justified.

Monoclonal Antibodies Currently Used for Cancer Immunotherapy and Their Mechanisms of Action

As of July 2013, 15 monoclonal antibodies (mAbs) are approved by the FDA and/or European Medicines Agency (EMA) for anticancer immunotherapy (Table 2). Catumaxomab, a rat/mouse hybrid bispecific antibody used for malignant ascites, is registered by the EMA, Health Canada and Israeli Ministry of Health, but not the FDA. This mAb binds via its Fc portion to an antigen-presenting cell, one combining site is directed to the transmembrane glycoprotein epithelial cell adhesion molecule (EpCAM), which is expressed on epithelial cancer cells, while the other site binds to CD3, a component of the T-cell receptor (TCR) complex on T lymphocytes.11,12 The consequent immunological reaction against neoplastic cells reduces the tumor burden from the abdomen of ascites cancer patients. Four of the mAbs nowadays used to treat cancer target the B-lymphocyte antigen CD20,13,14 which is expressed on 90% of B-cell non-Hodgkin's lymphomas, B-cell chronic lymphocytic leukemia (LCC), hairy cell leukemia and melanoma cancer stem cells but not on plasma blasts or plasma cells. Once the mAb ibritumomab binds its target, the radioactive label complexed by the chelator tiuxetan15 (Table 2) causes cell damage. With tositumomab, the attached ionizing radiation from 131I induces cell death.14-16 Moreover, for these 2 labeled mAbs as well as for rituximab14,17,18 and ofatumumab,14,19 other possible mechanisms of action include the activation of antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and CD20-mediated apoptosis.20 Cetuximab14,21,22 and panitumumab14,22 bind specifically to the epidermal growth factor receptor (EGFR) (also known as HER1, ErbB1) a type I receptor tyrosine kinase expressed by normal cells (e.g., of the skin and hair follicles) and tumor cells (head, neck, colon, rectum), hence competitively inhibiting the binding of ligands including the epidermal growth factor. This results in inhibition of cell growth, decreased production of vascular growth factor and pro-inflammatory cytokines and apoptosis. Pertuzumab, trastuzumab, and ado-trastuzumab emtansine all target HER2 (ErbB2).23 Trastuzumab inhibits the growth and proliferation of tumor cells that overexpress HER2,14,24 while ado-trastuzumab emtansine25 is the same mAb conjugated with the cytotoxic microtubule inhibitor DMI, arresting the cell cycle and leading to cell death. Pertuzumab inhibits the heterodimerization of HER2 with other ErbB receptors,26 blocking the mitogen-activated protein kinase (MAPK) and phosphoinositide-3-kinase (PI3K) signaling pathways and hence resulting in cell growth arrest and apoptosis.27 ADCC may also be involved in the cytotoxicity of this antibody. The antibody-drug conjugate (ADC) brentuximab vedotin is directed against CD30,28 a member of the tumor necrosis factor α (TNFα) receptor family and tumor marker expressed by activated T and B cells. Brentuximab vedotin arrests the cell cycle and induces apoptotic death owing to the monomethyl auristatin E (MMAE)-induced disruption of tubulin. By binding to the vascular endothelial growth factor (VEGF), bevacizumab29 prevents its interaction with fms-related tyrosine kinase 1 (FLT1, also known as VEGFR1) and kinase insert domain receptor (KDR, also known as VEGF2, CD309 and FLK1) on the surface of endothelial cells. This results in the inhibition of microvascular growth and metastatic disease. Alemtuzumab binds to CD52,30,31 which is present on mature lymphocytes, B cells, most monocytes and a number of other cells. The destruction of leukemic cells by this mAb mainly occurs by ADCC. Denosumab binds to tumor necrosis factor (ligand) superfamily, member 11 (TNFSF11, best known as RANKL),32 a transmembrane and soluble protein produced by osteoblasts and giant cell tumor of the bone (GCTB) cells that activates osteoclasts, the cells involved in bone resorption. RANKL-stimulated osteoclast activity is a mediator of the metastatic spread of cancer cells to the bone. Denosumab prevents the activation of RANKL on osteoclasts and osteoclast-like giant cells. Ipilimumab binds to cytotoxic T lymphocyte-associatd protein 4 (CTLA4) expressed on helper T cells. By interacting with its ligands CD80 (B7–1) and CD86 (B7–2), CTLA4 transmits an inhibitory signal that downregulate immune responses, unlike CD28, which also binds to CD80/CD86 but transmits a stimulatory signal. By blocking the interaction of CTLA4 with its ligands, ipilimumab therefore exerts an indirect T-cell mediated antitumor effect in melanoma patients.33

Table 2. Approveda monoclonal antibodies (mAbs) for cancer therapy.

Generic name
Type of mAb
Targetb
Mechanism of action
Approved indication(s)
Trade name
-omabs
Catumaxomab
Rat IgG2b / Mouse IgG2a bispecfic
EpCAMc/CD3d
Binds both EpCAM on tumor cell and CD3 on T cell
Malignant ascites
Removab®
Ibritumomab tiuxetane
Murine IgG1κ
CD20
Binds B cells and kills with ADCC,f CDCf and radiatione
Non-Hodgkin lymphoma
Zevalin®
Tositumomab-131I
Murine IgG2aλ
CD20
Binds to and kills B cells with 131I
Non-Hodgkin lymphoma
Bexxar®
-ximabs
Brentuximab vedoting
Chimeric IgG1κ
CD30h
Antimitotic MMAEg
Anaplastic large cell lymphoma; Hodgkin lymphoma
Adcetris®
Cetuximab
Chimeric IgG1κ
EGFRi
Binds to EGFR and turns off cell divisionj
Colorectal cancer; head and neck cancers
Erbitux®
Rituximab
Chimeric IgG1κ
CD20
Binds to CD20 on B cells leading to cell death
Non-Hodgkin lymphoma
MabThera® Rituxan®
-zumabs
Alemtuzumab
Humanized IgG1κ
CD52k
Eliminates lymphocytes
Chronic lymphocytic leukemia
Campath-1H®
Bevacizumab
Humanized IgG1κ
VEGFl
Angiogenesis inhibitor
Colorectal, lung, kidney, brain cancers
Avastin®
Pertuzumab
Humanized IgG1κ
HER2m
Inhibits dimeriz'n of HER2
with other HER receptors
Metastatic breast cancer
Perjeta®
Trastuzumab
Humanized IgG1κ
HER2
Prevents overexpression of HER2
Breast cancer
Herceptin®
Trastuzumab emtansinen
Humanized IgG1κ
HER2
mAb-drug conjugate. As for trastuzumab plus cytotoxic effect of mertansine (DM1)o
Advanced metastatic breast cancer
Kadcyla
-umabs
Denosumab
Human IgG2κ
RANKLp
Inhibits activation of osteoclasts by RANKL
Bone metastases; Giant cell tumor of the bone (GCTB)
Prolia®
Xgeva®
Ipilimumab
Human IgG1κ
CTLA-4q
Blocks interaction of
CTLA-4 with its ligandsr and enhances T cell activ'n
Metastatic melanoma
Yervoy®
Ofatumumab
Human IgG1κ
CD20
Binds to CD20 on B cell causing cell death
Chronic lymphocytic leukemia
Arzerra®
Panitumumab Human IgG2κ EGFRi Binds to and prevents activation of EGFR Colorectal cancer Vectibix®

International suffixes to distinguish the origins of therapeutic monoclonal antibodies: -omab, murine origin, usually IgG1 or IgG2; -axomab, bispecific mouse-rat hybrid mAbs (e.g., catumaxomab); -ximab, chimeric antibodies, usually IgG1, in which variable region is spliced into human constant region; -zumab, humanized antibodies with murine hyperviariable regions (usually IgG1) spliced into human antibody; -umab, antibodies from phage display or transgenic mice technology with a complete human sequence (IgG1 or IgG2). aApproved by FDA as at June 2013. bSpecificity of mAb. cEpCAM – epithelial cell adhesion molecule. Expressed on epithelial and epithelial-derived neoplasms. dCD3 – part of the TCR complex on T lymphocytes. eWith Yttrium-90 or Indium-111. Tiuxetan is a chelator. fADCC – antibody-dependent cell-mediated cytotoxicity; CDC – complement-dependent cytotoxicity. gConjugated to the cytotoxic agent monomethyl auristatin E (MMAE). hCD30 or TNFRSF8, a cell membrane protein of tumor necrosis receptor family. Expressed on activated T and B lymphocytes. iEGFR – epidermal growth factor receptor (HER1, ErbB-1). jNot effective in cases with KRAS mutations in cancer cells. kPresent on the surface of mature lymphocytes and associated with some types of lymphomas. lVEGF – vascular endothelial growth factor (a subfamily of growth factors; includes VEGF-A). mHER2 – human epidermal growth factor receptor 2. Also known as Neu, ErbB2, CD340 or p185. nmAb linked to the cytotoxin mertansine (DM1). In the US known as ado-trastuzumab to distinguish from trastuzumab. oA maytansinoid tubulin inhibitor. pRANKL - receptor activator of nuclear factor kappa-B ligand (CD254), a member of the TNF cytokine family. qCTLA-4 – cytotoxic T-lymphocyte antigen 4; CD152. rLigands for CTLA-4 – CD80/CD86.

Range of Side Effects of Monoclonal Antibodies

Although mAbs used for cancer immunotherapy are generally better tolerated than widely used ‘conventional’ chemotherapeutic agents, adverse events following the administration of mAbs can result from a variety of mechanisms and may be quite diverse. Table 3 summarizes the most frequently occurring/important systemic and cutaneous adverse events of the 15 different mAbs currently used in cancer treatments, summarizing information from published medical/scientific literature; research studies; reports of clinical trials; FDA prescribing information, warnings, precautions, and adverse reactions; data released by drug companies; and prescribing information revisions as a result of post-marketing experience. The wide variety of mAB-associated reactions ranges from, for example, headache, mild gastrointestinal symptoms such as diarrhea, transient rash and itching to severe cytopenias, cardiac toxicity, anaphylaxis, exfoliative dermatitis and rarely life-threatening bullous toxidermias. Being non-endogenous proteins of sufficient size, immunogenicity is always a safety concern and despite progressive efforts in developing chimeric, humanized, and fully human mAbs, the possibility of generating anti-idiotype antibodies indicated that the potential immunogenicity of mAbs persists, at least to some degree.70 Although ADRs such as anaphylaxis, serum sickness, autoimmune diseases, urticaria, Stevens-Johnson syndrome (SJS), and toxic epidermal necrolysis (TEN) are clearly mediated by the immune system,8 others like some cytopenias (thrombocytopenia,71,72 neutropenia73,74 and anemia75) may or may not be so. Some pulmonary76 and liver77,78 toxicities, induced infections and cutaneous responses79-81 are often less well defined and understood and may have at least an indirect connection to immunological processes. This might also be said about IRs/CRS and the systemic inflammatory response syndrome (SIRS),82-85 but the tumor lysis syndrome (TLS)86 and some cytopenias,72,87 as well as heart, pulmonary, hepatic, kidney, embryo-fetal, and neurological toxicities appear to be due to direct cytotoxic actions and/or a number of other non-immune mechanisms.88,89

Table 3. Hypersensitivity/adverse reactions of monoclonal antibodies used for cancer therapy.

mAbsa Hypersensitivityb/adversec reactions
Refs
Systemic Cutaneous
Catumaxomabd
SIRS; abdominal disorders; CRS; pyrexia; cytopeniase; hepatotoxicity; dyspnoea; infections; immunogenicity
Rash; erythema; hyperhidrosis; pruritus; allergic dermatitis
34 , 35
Ibritumomab
tiuxetan
IR; infections; severe cytopenias; immunogenicity; secondary malignancies
EM; SJS; TEN; bullous dermatitis; exfoliative dermatitis
36
Tositumomab-I131
Anaphylaxis; severe cytopenias; IR; fetal harm; hypothyroidism; secondary malignancies; infection; immunization
In clinical trial: Skin reactions, all grades - rash 17%, pruritus 10%, sweating 8%. Grades 3 and 4 - 0 - < 1%; exfol. dermatitis
37 , 38
Brentuximab
vedotin
PN; IR; cytopenias; TLS; immunogenicity; PML; fetal harm; anaphylaxis
SJS; rash; pruritus; alopecia
39 , 40
Cetuximab
IR; cardiopulmonary arrest; pulmonary toxicity; electrolyte imbalance; GI; infection
Acneiform rash; nail changes; pruritus; xeroderma; paronychial inflammation
18 , 41 44
Rituximab
IR; TLS; PML; renal toxicity; infections;
cardiac events; pulmonary events; bowel obstruct. and perforation; neutropenia; RA; anaphylaxis; HBR; SS
Paraneoplastic pemphigus; lichenoid dermatitis; vesiculobullous dermatitis; SJS; TEN
18 , 44 50
Alemtuzumabf
IR; cytopenias; infections,g immunogenicity; cardiac events; pulmonary events
Urticaria; rash; erythema; pruritus
44 , 51 53
Bevacizumab
GI perforation; hemorrhage; wound healing complications; thrombosis; IR; hypertension; necrotizing fasciitis; proteinurea; pulmonary events
Exfoliative dermatitis; alopecia
44 , 53 57
Pertuzumab
Embryo-fetal toxicity; IR; cytopenias; GI; PN; hypersensitivity/anaphylaxis; LVD
Alopecia; rash; paronychia; pruritus
58 , 59
Trastuzumab
Cardiomyopathyh; embryo-fetal toxicity; IR; LVD; pulmonary eventsi; neutropeniaj; anaphylaxis/angioedema; anemia; GI
Rash; nail disorders; pruritus
18 , 44 , 60 ,
61
Trastuzumab
Emtansinek
Hepatotoxicity; LVD; fetal harm; pulmonary events; thrombocytopenia; neurotoxicity; hypersensitivity/IR
Rash; pruritus
62
Denosumab
Hypocalcemia; embryo-fetal toxicity; ONJ and osteomyelitis; fatigue/asthenia; dyspnea
Dermatitis; eczema; rash; pruritus
63
Ipilimumab
IMR; diarrhea; fatigue
Dermatitis; pruritus; rashl
64 66
Ofatumumab
IR; cytopenias; intestinal obstruction; PML; HBR; pneumonia; pyrexia; infections; cough; dyspnea; diarrhea; fatigue
Rash; urticaria; hyperhidrosis
67 , 68
Panitumumabm,o IR; pulm. Fibrosisn; pulmonary embolism, electrolyte depletion; GI; fatigue Rash; dermatitis 'acneiform'; exfoliation; erythema; pruritus; xerosis; paronychia; skin fissures; photosensitivityo 18 , 69

a Approved by FDA and/or European Medicines Agency (EMA). bReactions known or suspected of having an immunological basis. cOther adverse reactions with no clearly established, or yet to be demonstrated, immune mechanism (eg.nausea, cough, diarrhea, fatigue, sweating etc). dRegistered by EMA, Health Canada and Ministry of Health, Israel but not FDA. eMeans one or more of anemia, lymphopenia, neutropenia, thrombocytopenia. fWithdrawn from US and Europe in 2012 to be re-launched for MS. gIn particular, Pneumocystis jiroveci, CMV, EBV, herpes virus. hLeft ventricular dysfunction. Greatest risk when administered with anthracyclines. iInterstitial pneumonitis, dyspnea, ARDS, pulmonary infiltrates/fibrosis/edema, pleural effusions. jHighest with myelosuppressive therapy. kCalled Ado-trastuzumab emtansine in the US lSJS/TEN, Sweet's syndrome, DRESS seen rarely. mNot indicated for use in combination with chemotherapy due to increased toxicity. nShould be discontinued in patients developing interstitial lung disease, pneumonitis, lung infiltrates. oMost common adverse reactions are skin toxicities. CRS, cytokine release syndrome; EM, erytheme multiforme; GI, gastrointestinal symptoms, e.g., nausea, diarrhea, vomiting, constipation etc; HBR, hepatitis B reactivation; IMR, immune-mediated reactions due to T cell activation and proliferation – enterocolitis, hepatitis, dermatitis, neuropathies, endocrinopathies; IR, infusion reactions; LVD, left ventricular dysfunction; ONJ, osteonecrosis of the jaw; PML, progressive multifocal leukoencephalopathy; PN, peripheral neuropathy; RA, rheumatoid arthritis; SIRS, systemic inflammatory response syndrome; SJS, Stevens-Johnson syndrome; SS, serum sickness-like reactions; TEN, toxic epidermal necrolysis;, TLS, tumor lysis syndrome;

mAb-induced ADRs with an apparent immune basis, those with an indirect immune connection, reactions with a possible—even if minor—immune component and adverse events caused by non-immune toxicities are discussed below.

Hypersensitivity/Adverse Reactions to Monoclonal Antibodies

These are adverse responses that fall into at least one of the 4 categories of hypersensitivity, Types I, II, II and IV, as defined by Gell and Coombs5 (Table 1). MAbs are known to cause all 4 types of hypersensitivities.

Type I (immediate) hypersensitivity

These reactions, mediated by IgE antibodies and sometimes called anaphylactic hypersensitivities, are relatively uncommon upon the administration of mAbs. Immediate hypersensitivity may affect a single organ such as the nasopharynx (allergic rhinitis), eyes (conjunctivitis), mucosa of mouth/throat/tongue (angioedema), bronchopulmonary tissue (asthma), gastrointestinal tract (gastroenteritis), and skin (urticaria, eczema), or multiple organs (anaphylaxis), causing symptoms that range from minor itching and inflammation to death. Anaphylactoid or pseudo-allergic reactions may also provoke flushing, hypotension, mucous secretion, rash, pruritus, and urticarial, making it difficult to distinguish these symptoms from a true, IgE-mediated Type I hypersensitivity reaction, which is generally accompanied by similar, though more severe, symptoms.8 In particular, cardiovascular collapse and bronchospasm occur frequently in the course of anaphylaxis while cutaneous manifestations are seen often during anaphylactoid reactions. Because of the immunogenic potential of the mAbs mentioned above, especially chimeric molecules (brentuximab, cetuximab, rituximab) and those that contain murine or rat sequences (catumaxomab, ibrutumomab, tositumomab), mAbs commonly carry warnings of possible immediate reactions including anaphylaxis, but actual incidence of such reactions is quite small. Anaphylaxis has been reported for cetuximab, rituximab, trastuzumab, pertuzumab, tositumomab, and ibritumomab, and these latter two mAbs have also been shown to cause bronchospasm and angioedema.36,37,41,45,58,60,90-94 Cetuximab-reactive IgE antibodies found in the sera of some patients with anaphylaxis to this drug were found to be specific for the disaccharide α-D-galactose-(1–3)-β-D-galactose present at asparagine 88 of the heavy chain of the Fab fragment.92 Most patients who reacted already had the antibodies in their serum before receiving the drug. The analysis of patients treated with rituximab at Massachusetts General Hospital in 2006–2010 showed that 79 of 901 patients (9%) experienced an immediate hypersensitivity reaction with most (76%) developing symptoms during their initial infusion.95 The authors concluded that immediate hypersensitivity to rituximab commonly occurs after the initial infusion and found that Waldenstom's macroglobulinemia patients had a disproportionately higher risk of hypersensitivity. Nonetheless, no evidence of rituximab-specific IgE antibodies or positive skin tests to the mAb were presented.

Infusion reactions or cytokine release syndromes

The infusion of mAbs typically provokes a characteristic infusion syndrome, usually occurring within a few hours from the beginning of the administration. Most reactions are typically mild to moderate with 'flu'-like symptoms of fever, chills, rigors, nausea, headache, asthenia, rash, pruritus, etc...96 In a small percentage of patients developing potentially fatal reactions, mostly with in the course of the first or second infusion, some signs and symptoms such as hypotension, cardiac arrest, bronchospasm and urticaria may be common to a true Type I, IgE antibody-mediated anaphylactic reaction, making it difficult to discriminate between these two events,8,97 although IgE-mediated reactions generally have a faster and more severe onset (usually within minutes). IRs, including severe ones, have been reported for all mAbs, the incidence of such reactions (e.g., in response to cetuximab) being similar to that range seen with taxanes (~20–40%) and platinum derivatives (~12–16%).97 Rituximab and trastuzumab induce the highest incidence of IRs. In general, the incidence of mAb-induced IRs varies from ~15–20% for cetuximab (3% grade 3–4) and 40% for trastuzumab first infusion (< 1% grade 3–4) to 77% for rituximab first infusion (10% grade 3–4). Even after the fourth infusion, 30% of cancer patients react to rituximab, and the incidence of IRs remains of 14% after the eighth infusion. Approximately 80% of fatal reactions occurred after the first rituximab infusion. The incidence of IRs to the humanized mAb bevacizumab and the fully humanized agent panitumumab are significantly lower: < 3% (0.2%) for the former and 3% (~1%) for the latter.97 The mechanisms underlying mAb-induced IRs remain incompletely understood. It is thought that the mAb-target interaction may lead to release of cytokines such as TNFα and interleukin (IL)-6 that can produce a range of symptoms seen in infusion reactions, some of which being similar to those seen in Type I allergic responses. Some results indicate that the severity of IRs is related to the number of circulating lymphocytes, for example, a lymphocyte count > 50 x 109/L was likely to be associated with a severe reaction,98 and the regression of IR symptoms in oxaliplatin-treated colorectal cancer patients paralleled a decrease in serum cytokine concentrations.99 Note that even repeated severe IRs to a particular mAb may not necessarily preclude the administration of another appropriately targeted antibody as demonstrated by the safe use of 131I-tositumomab after repeated IRs to rituximab.100

Are some adverse reactions to mAbs true Type II or Type III hypersensitivity responses?

A number of case reports and retrospective analyses suggest that some mAbs, principally rituximab, may cause Type II antibody-mediated cytotoxic hypersensitivity reactions. However, the oncology and some other medical specialty literatures do not often use this terminology, which is predominantly allergy- and immunology-based. Thrombocytopenia, neutropenia and anemia can occur in some patients treated with mAbs as part of anticancer immunotherapy, but the mechanisms of these potentially serious side effects frequently remain unexplored. Autoimmune forms of thrombocytopenia and hemolytic anemia are Type II hypersensitivities. They are well known complications of lymphoproliferative diseases and occur predominantly in patients with lymphocytic leukemias.101 For example, late-onset neutropenia, especially after rituximab treatment, has been examined in a growing number of reports but with each of the 3 cytopenias seen during mAb therapy, it is frequently unclear whether or not the depletion of cell numbers is due to an immunological mechanism, i.e., whether it represents a true Type II hypersensitivity response. Likewise, Type III hypersensitivities such as serum sickness-like reactions and vasculitis are also known to occur in response to mAbs, but their frequency and extent during cancer immunotherapy is likely to be underestimated. Some pulmonary events, including mAb-induced lung diseases, are hypersensitivity reactions that result from interaction of the drug with the immune system and involve drug-specific antibodies or T cells. The possible involvement of mAbs in each of these drug-induced Type II and Type III reactions is discussed below.

mAb-induced thrombocytopenia

Compared with the incidence of thrombocytopenia in patients given chemotherapy, grade 3/4 thrombocytopenia to rituximab monotherapy is seen in only 1.7% of patients102 and this figure is barely increased when the mAb is combined with chemotherapy. Cattaneo et al.103 reported an incidence of 10.4% in 72 patients affected by non Hodgkin’s lymphoma (NHL) and given a total of 317 ritiximab infusions. Transient severe acute thrombocytopenia after rituximab in one patient with hairy cell leukemia and another with mantle cell lymphoma was reversible in a few days following withdrawal of the antibody, but the underlying mechanism remained unclear. While acknowledging that the expression of CD20 (the target of rituximab) on platelets and their rituximab-dependent immune-mediated lysis might have contributed to this event, the authors pointed out a massive bone marrow involvement of neoplastic B lymphocytes.104 From the study of a case of thrombocytopenia induced by rituximab and a review of the literature, Ram et al. failed to deduce the presence of rituximab-dependent antibodies and found that the levels of IL-1 and IL-6 were not increased but those of complement were. They concluded that mAb-induced transient thrombocytopenia might be associated with a CRS and is probably mediated by complement activation. A severe case of potentially life-threatening thrombocytopenia and at least 2 other cases induced by the HER2-targeted mAb trastuzumab have been reported.61,105,106 About 3% of patients given alemtuzumab for early multiple sclerosis developed potentially fatal thrombocytopenia107,108 and this mAb was shown to provoke lymphopenia, neutropenia and thrombocytopenia in 5 of 11 patients with peripheral T-cell lymphoproliferative disorders.109 In this setting, lymphopenia may be due to the direct targeting of CD52 on lymphocytes. Bevacizumab (which target VEGF) has been associated with arterial thromboembolism110 and it may facilitate venous thromboembolic events.111

mAb-induced neutropenia

Rituximab is associated with both early and late neutropenia and it seems these are caused by different mechanisms. In early studies in the 1990s, grade 3/4 neutropenia was reported in 4.2% of patients given rituximab.102 It is now known that late onset neutropenia (LON), that is neutropenia occurring at least 4 weeks after administration of the mAb, has a comparatively higher incidence, with 8 different studies revealing figures of from 4% to 27.3%.112 Some claim the prevalence of this reaction is underestimated.103 While the mechanisms underlying LON are poorly understood, results suggest that direct cytotoxicity is unlikely47 (as CD20 is not expressed on granulocytes and progenitor cells) and currently no single explanation has universal support. Immune-mediated mechanisms have been advanced for rituximab-induced LON, including the production of autoantibodies113 and the expansion of large populations of granular lymphocytes leading triggering neutrophil apoptosis upon the activation of Fas and other interactions.114-116 Dunleavy et al.112 have proposed an interference with the neutrophil emigration from the bone marrow driven by stromal-derived factor-1 (SDF-1), a chemokine required for the development of early B-cell and granulocyte precursors in the bone marrow, while Terrier et al.117 believe there is hematopoietic lineage competition due to excessive B-cell activating factor (BAFF)-induced B-cell recovery over granulopoiesis.

mAb-induced hemolytic anemia

Severe anemia has been reported in 1.1% of patients on rituximab monotherapy,102 but one study showed an incidence of 5.2%.103 Some notable studies report the development of severe autoimmune hemolytic anemia (AIHA) in a patient with a lymphoproliferative disorder,118 as well as cases of intravascular hemolysis119 and multiorgan ischemia due to an autoimmune anti-Pr cold agglutinin.120 Besides rituximab, alemtuzumab has been to provoke pure red cell aplasia and AIHA,121,122 exacerbate pre-existing AIHA123 and has been involved in the death of a patient due to refractory AIHA.124 Bevacizumab has been implicated in cases of renal thrombotic microangiopathy.125,126

mAb-induced vasculitis

Hypersensitivity vasculitis induced by drugs is a manifestation of a Type III response. Drug-induced vasculitis (DIV) usually occurs in the skin and sometimes in subcutaneous tissue, kidneys, and the lungs. Cutaneous vasculitis (CV), a small vessel systemic vasculitis manifesting as palpable purpura, ranges in severity from benign and self-limiting to life-threatening with multiorgan failure. The mechanisms underlying DIV are still incompletely understood but cellular as well as humoral immune processes appear to be involved. Although CV associated with the TNFα inhibitor infliximab in the treatment of arthritis is well known,127 this condition is rarely seen following mAb-based cancer therapy. Rituximab-induced vasculitis is the subject of a number of reports128,129 including a case of CV induced by a first infusion in a patient with B-cell chronic lymphoid leukemia (CLL) confirmed by histological samples that displayed the typical features of small vessel leukocytoclastic vasculitis. No expression of CD20 was detected on blood vessel walls, making a specific interaction of the mAb with vessel walls unlikely.49 Although the number of cases is still relatively small, it seems that vasculitis may be associated with cases of CLL.

mAb-induced serum sickness-like reactions

Serum sickness, a classical Type III hypersensitivity reaction due to protein antigen-antibody complexes, occurs as a response to foreign antigens such as antitoxins, antivenins, and vaccines. Although non-protein antigens generally do not provoke the response, some drugs induce a reaction that is clinically similar.8 Symptoms, which typically appear 6–21 d after drug administration, include lymphadenopathy and fever. Cutaneous symptoms, often urticarial and morbilliform eruptions and sometimes erythema and petechiae, occur in up to 95% of patients. Angioedema, arthritis, arthralgia, and gastrointestinal symptoms are common, joints may be severely affected and splenomegaly, hepatomegaly, peripheral neuropathies, pericarditis, and encephalomyelitis are seen. Chimeric mAbs have the potential to induce serum sickness and this has in fact occurred, the first of such examples being a reaction to rituximab reported in 2001.130 Recently, it has been claimed that rituximab-induced serum sickness-like reactions occur in 1–20% of patients,48 more commonly in patients with autoimmune diseases (particularly autoimmune thrombocytopenia) and marked hypergammaglobulinamia, and it has been proposed that these two conditions are predisposing factors for the development of the reactions.131,132

Autoimmune diseases

Autoimmune diseases caused by mAbs are rare but one mAb used for cancer therapy, namely, ipilimumab (targeting CTLA4) has been shown to operate as an immunostimulatory agent by CTLA4 blockade and increased T-cell stimulation133,134 to produce an autoimmune enterocolitis and/or a range of other adverse reactions including rash and hepatitis.135

Pulmonary adverse events caused by mAbs

Pulmonary adverse events caused by mAbs comprise a heterogeneous group of lung diseases often classified under the title drug-induced lung diseases (DILD). Since the mechanisms underlying such lung injuries have generally not been worked out, any classification on the basis of pathogenesis is difficult. Adverse events can be grouped into 4 main categories: interstitial pneumonitis and fibrosis; acute respiratory distress syndrome (ARDS), bronchiolitis obliterans organizing pneumonia (BOOP), and hypersensitivity reactions; although, a range of different classifications may be seen in the literature. Reports of hypersensitivity pneumonitis, a combined Type III and IV hypersensitivity reaction in a TH1/TH17 response,136 are increasing, particularly upon the administration of antineoplastic drugs. Table 4 lists the mAbs used for cancer therapy that are the main offenders in provoking pulmonary adverse reactions, and shows the different individual adverse reactions induced by each mAb. Once again, rituximab is the most implicated mAb, inducing a heterogeneous spectrum of lung disorders44,50,137-146 In 2003, the reported rate of possible drug-induced lung injury was < 0.03% from > 540,000 exposed patients.138,147 BOOP is the most common clinical diagnosis of rituximab-induced lung disease, followed by interstitial pneumonitis, ARDS and hypersensitivity pneumonitis.137 In a review of 45 cases of lung disease ascribed to this mAb, 3 groups of patients were identified on the basis of the time of onset of the reactions.50 The most common group exhibited acute/subacute hypoxemic organizing pneumonia starting 2 weeks after the last infusion (i.e., early onset); ARDS occurred within a few hours usually after the first infusion; and organizing pneumonia developed long after rituximab therapy. Based on a number of factors including the recurrence and increasing severity of symptoms with the number of infusions administered, the responsiveness to steroid therapy, rash, eosinophilia, and the occurrence of lymphocytes within the bronchoalveolar lavage (BAL) fluid, the authors suggested that early onset organizing pneumonia reflects a hypersensitivity reaction to the chimeric antibody. While late onset organizing pneumonia was thought to be either related to mAb toxicity or to immune system restoration, the commencement of ARDS symptoms within a few hours of infusion suggested either a CRS or a TLS, with no relationship to hypersensitivity. The pathogenesis of rituximab-induced interstitial lung disease remains largely unknown but complement activation and cytokine secretion, particularly of pro-inflammatory TNFα, may be involved.148 ARDS reported after a single infusion of rituximab has also been associated with release of pro-inflammatory cytokines.146 A case of fatal intra-alveolar hemorrhage with appearances of hypersensitivity pneumonia (including elevated eosinophil count and serum IgE levels) has been reported upon rituximab administration. The authors of this report suggested that the pathogenesis of such reactions may be due to activation of cytotoxic T lymphocytes, resulting in vascular and alveolar damage, persistent cytokine release and a possible cross-reaction between lung and tumor antigens.149

Table 4. Pulmonary adverse events caused by monoclonal antibodies (mAbs) used in the treatment of cancersa.

mAb Pulmonary adverse events
Cetuximab
Interstitial pneumonitis
Rituximab
ARDS
BOOP
Bronchospasm
Diffuse alveolar hemorrhage
Hypersensitivity pneumonitis
Interstitial pneumonitis
Alemtuzumab
Bronchospasm
Diffuse alveolar hemorrhage
Pulmonary infections (e.g., TB, aspergillosis)
Bevacizumab
Bronchospasm/anaphylaxis
Pulmonary hemorrhage from site of tumor
Trastuzumab
ARDS
BOOP
Interstitial pneumonitis
Panitumumabb Interstitial lung diseaseb
Lung infiltrates
Pneumonitis
Pulmonary fibrosis

a Data from ref. 44bDiscontinue panitumumab in patients developing interstitial lung disease.69 ARDS, acute respiratory distress syndrome; BOOP, bronchiolitis obliterans organizing pneumonia

Delayed-type (Type IV) hypersensitivity reactions to mAbs

Unlike Type I, II, and III hypersensitivities that are mediated by antibodies, Type IV reactions depend upon antigen-specific TH1/TH2/TH17 lymphocytes and effector mechanisms involving the activation of cytotoxic lymphocytes, macrophages, and eosinophils. Type IV cutaneous reactions to drugs generally become apparent 7–21 d after exposure, with reactions reaching a maximum after 24–72 h. Subsequent reactions may develop only within 1 or 2 d. Sensitivity to the provoking antigen can be transferred by lymphoid cells.5,8 Examples of drug-induced Type IV cutaneous reactions are allergic contact dermatitis, maculopapular exanthema, psoriasis, acute generalized exanthematous pustulosis (AGEP), fixed drug eruption (FDE), drug reaction with eosinophilia and systemic symptoms (DRESS), SJS, TEN, and erythema multiforme (EM). These different reactions are associated with different T-cell subsets, each of which secrete a peculiar, yet not mutually exclusive, profile of cytokines and chemokines.8,150 Type IV cutaneous hypersensitivity reactions induced by mAbs used in cancer therapy are rare, with reports seemingly confined mainly to ibrutumomab, brentuximab, and rituximab (Table 3). Cases of dermatitis induced by some of the other mAbs, including catumaxomab, tositumomab, bevacizumab, denosumab, ipilimumab, and panitumumab, may in fact be true Type IV responses as the underlying mechanisms have rarely been investigated (see below). The incidence of serious mucocutaneous reactions in 125,000 patients given rituximab between 1997 and 2001 was estimated to be 0.0008%, which is lower than the expected incidence of such reactions in lymphoma patients.90 In fact, cutaneous side effects to rituximab that are not usually serious, are fairly often seen, and generally occur 1 to 13 weeks after drug withdrawal. However, SJS, TEN lichenoid dermatitis, vesiculobullous dermatitis, and paraneoplastic pemphigus in response to this agent have been reported.90,151,152 It has been suggested that the rarely occurring SJS may be associated with cases of follicular NHL.131

Target-related mucocutaneous reactions to mAbs

Examples of such reactions which are not genuine hypersensitivities (i.e., not immune-mediated) include adverse reactions provoked by administration of agents that bind EGFR. As with tyrosine kinase inhibitors such as gefitinib and erlotinib, cetuximab and panitumumab commonly cause a moderate to severe, dose-dependent so-called “acneiform” or papulopustular eruption in 50–100% of patients.153 Still, such a reaction is more severe and widespread with EGFR-targeting mAbs than with small EGFR inhibitors. In the main, the eruption is confined to seborrheic regions, namely, the face, scalp, neck, shoulders, and upper trunk. EGFR is expressed in the epidermis, sebaceous glands, and hair follicles, where it is thought to have an important function in maintaining the health of the epithelium. Although the mechanism of EGFR-targeted mAb-induced skin rash is incompletely understood, it is speculated that inhibition of EGFR causes alterations and rupture of the epithelial barrier, allowing for bacterial access and proliferation, and ultimately the characteristic rash.153,154 In addition to the papulopustular rash, other less frequent adverse effects include xerosis, palmar-plantar rash, pruritus, talangiectasia, fissures, nail changes (e.g., paronychia), hair changes (e.g., trichomegaly, alopecia), hyperkeratosis, mucositis, pyrogenic granuloma, and hyperpigmentation of the skin.154,155

Cardiac adverse events

Cardiac adverse events have occurred with at least 4 of the mAbs approved for cancer therapy. Cardiopulmonary arrest and/or sudden death resulted in ~2% (4) of 208 patients given cetuximab and exposed to radiation therapy. No reactions were seen in 212 patients given radiation alone.41 Cardiac arrhythmias have been reported for rituximab,45 cardiomyopathy following trastuzumab,60 and left ventricular dysfunction (LVD) for pertuzumab, trastuzumab, and trastuzumab emtansine.58,60,62 Decreases in left ventricular ejection fraction may occur with drugs that block HER2 (ErbB2) activity such as pertuzumab and trastuzumab and it appears that pertuzumab-treated patients given anthracyclines or radiotherapy to the chest may be at higher risk of decreased LVD. Patients receiving trastuzumab alone or in combination therapy show a 4–6-fold increase in the incidence of myocardial dysfunction and again, the incidence is highest when the mAb is administered in combination with an anthracycline. Animal models have been developed to investigate the mechanism of trastuzumab-induced cardiac dysfunction.18,156 Mice with cardiac-specific deletion of ERBB2 develop cardiomyopathy and sensitivity to anthracyclines with age157 and trastuzumab has been found to inhibit neuregulin 1 (NRG1), a growth factor necessary for cardiac development and maintenance of heart structure and integrity.18,158

Tumor lysis syndrome

TLS occurs 48–72 h after the start of anticancer chemotherapy as a result of the rapid, drug-mediated death of large numbers of malignant cells. This results in hyperkalemia, hypercalcemia, hyperphosphatemia, and hyperuricemia, a profound ionic imbalance and a possible progression to acute renal failure, cardiac arrhythmias, seizures, and death.159 TLS, unlike CRS, is not difficult to distinguish from Type I immediate hypersensitivity reactions. TLS is seen most often in patients with leukemias and high-grade lymphomas and, apart from small-cell cancer and neuroblastoma, only rarely in association with solid tumors. The syndrome is well known after brentuximab vedotin and rituximab (incidence ~0.1–0.15%), especially in patients with high tumor load, but the reaction elicited by rituximab is somewhat an atypical TLS and it remains incompletely characterized.90

Progressive multifocal leukoencephalopathy

PML is a progressive, usually fatal viral disease that in some respects resemble multiple sclerosis, as the myelin sheath of nerve cells is ultimately destroyed affecting transmission of nerve impulses.160 It occurs in severely immunodeficient individuals, e.g., in transplant patients on immunosuppressants or AIDS patients, but it is also occasionally seen upon the administration of mAb directed to B cells, in particular, brentuximab, rituximab, and ofatumumab. The infective agent implicated, the so-called JC virus, is a member of Polyomaviridae that persists asymptomatically in about one third of the population. In 2009, 57 cases of PML after rituximab therapy in HIV-negative patients were reported.161 Earlier, the labeling of the mAb had been amended to indicate the risk of infections, including infections with JC virus.

Need for Systematic Testing to Accurately Identify True Hypersensitivity Reactions to Monoclonal Antibodies

True hypersensitivity reactions have an immunological basis, be it humoral and/or cell-mediated,5,8 the mechanisms of an adverse event are not always clearly established before calling it a hypersensitivity response. The scientific literature, clinical trials reports, information provided by drug companies, package inserts, and several other sources of information are replete with examples of such an etymological misuse. There are a number of reasons for this situation. First, a widely accepted definition of a hypersensitivity reaction is lacking and confusion is generated even within its “home” disciplines of immunology and allergy.162 Moreover, the sheer range and variety of possible drug-induced reactions include cytopenias, vascular disorders, liver injury, lung diseases, and many mucocutaneous reactions, de facto making it difficult to reach a definitive classification. Skin prick and intradermal testing can often, but not always,8 be used to detect immediate Type I and delayed Type IV reactions to drugs, and results obtained with antigens such as proteinaceous mAbs should be reliable if the tests are properly performed.8 True Type I reactions are mediated by IgE antibodies and immunoassays to detect IgE specific for individual mAbs are not difficult to devise and develop. Patch testing is both a screening test for hypersensitivity and a provocation test in the skin8 and can be used to investigate delayed cutaneous reactions.

Systemic reactions

Drug-induced thrombocytopenia, neutropenia, and anemia may be the result of an immune-mediated or cytotoxic mechanism, while liver and lung injuries can show multiple and varied manifestations that appear to be the result of either an immune or non-immune mechanism. This difficulty in assessing and sorting out the signs and symptoms of ADRs is compounded by the absence of well-established clinical and laboratory markers and appropriate tests. Distinguishing between drug-induced thrombocytopenia, neutropenia, and anemia that is either an immune-based suppression of hematopoietic cell lines or a dose-dependent bone marrow cytotoxicity is not always easy, especially in the case of chemotherapeutics, which are often taken as part of combinatorial regimens and for which the marrow suppression of megakaryocytopoiesis is a well-recognized side effect. Although some in vitro tests that detect platelet-reactive serum antibodies are available to aid the diagnosis of drug-induced thrombocytopenia,163,164 the tests are not standardized and sometimes involve technical difficulties (e.g., related to drug solubility and to metabolic conversion). In addition, the facts that tests are sometimes available only in a few reference laboratories and that results are not immediately available in urgent situations are major drawbacks. Likewise, although some anti-neutrophil antibody tests are used to help in the diagnosis of immune-mediated neutropenia and agranulocytosis,73,74,165 these assays are not widely available, technical difficulties can be limiting (for instance, Fc receptors on neutrophils can lead to false-positive results) and findings may not always be easy to interpret. At first sight, the tests for drug-induced anemias are not as big a problem, but sorting out the different mechanisms involved, including (but not limited to) IgG/IgM to drug-cell membrane complex, complement activation, and the presence of autoantibodies that may be drug-dependent or independent and may react either in the presence or absence of the drug, is often a challenging task.75 For drug-induced vasculitis there are no markers that can be confidently used to distinguish the condition from other vasculitis,166 and making a confident diagnosis of immune-mediated drug-induced liver injury (DILI) remains difficult, again due to the absence of reliable specific tests.77 The situation with DILD is similar: as in vivo drug provocation tests are judged too risky and lymphocyte transformation tests have been applied but appeared to be inadequate,167 high resolution CT scanning, pulmonary function testing, and bronchoscopy with BAL are often relied upon along with the patient's drug exposure history and cause exclusion.168,169

Mucocutaneous reactions

Type IV hypersensitivity reactions of the skin and mucous membranes seen during and after mAb administration such as contact dermatitis, maculopapular rash, AGEP, and lichenoid rash can be investigated by patch testing but this test appears to be less useful for SJS, TEN, and DRESS, for which the timing of the evaluation is important. ELISPOT cytokine assays for the detection of drug-reactive T cells, tests for cutaneous lymphocyte-associated antigen (CLA) as well as the monitoring of the skin-associated chemokine (C-C motif) ligand 27 (CCL27) and its interaction with chemokine (C-C motif) receptor 10 (CCR10) are other promising diagnostic approaches.8 Methods to identify the mechanisms of some drug-induced local toxicities are not as obvious. This applies to reactions involving the mouth, digestive tract, scalp, hands, feet, nails, and hair, often manifesting as inflammation and ulceration of mucous membranes, rashes, skin fissures, xerosis, some photosensitivities, nail dystrophies, pigmentary changes, pruritus, etc. At least some of these responses are in need of close investigation to establish whether or not any immune processes are involved during any stage of the reaction. This is particularly relevant for targeted therapies that are associated with poorly characterized adverse reactions of mucous membranes, skin, nails, and hair, such as EGFR inhibitors.79-81

Concluding Remarks: Do Some Hypersensitivity Responses to Monoclonal Antibodies go Unrecognized?

MAbs have revolutionized the treatment of cancers. Besides their antineoplastic effects, these agents are associated with less adverse reactions than conventional chemotherapy, and these tend to be better tolerated. Nevertheless, a wide spectrum of adverse events to mAbs is observed, necessitating efforts to minimize side effects and to identify, describe and manage the reactions. In any assessment of the side effects of a drug, the possibility of hypersensitivity reactions is always considered and reference to the appearance or absence of such responses is commonly found in clinical trial reports, information released by regulatory agencies and pharmaceutical companies, toxicological investigations and the immunology, pharmacology, and oncology literature. Although “hypersensitivity” is widely used across a number of disciplines, there appears to be no common agreement on the definition of this term, as demonstrated by the sometimes different features of responses called hypersensitivities. Even though existing testing procedures to precisely identify true drug-induced Type I, II, III, and IV hypersensitivities are frequently inadequate, the systematic and more widespread application of the tests that are available would undoubtedly improve the discrimination of many hematological, vascular, lung, liver, and cutaneous adverse events, leading to an improved analysis of many reactions as well as to an accurate identification of those with an immunological/allergic basis. Further research is needed to refine and standardize existing tests, make them widely accessible and to develop new and improved procedures for the elucidation of mechanisms and accurate diagnosis. This area of chemotherapy has been neglected, especially by immunologists and allergists, such that the true nature of many adverse events remain imprecisely defined. Nowhere this is more apparent than with those drug-induced reactions that are now sometimes incorrectly called hypersensitivities or mistakenly classified as being non-hypersensitivity responses.

Disclosure of Potential Conflicts of Interest

No financial contribution or any other assistance was received by the author in relation to the preparation and presentation of this work. No potential conflict of interest is, or has been, involved.

Glossary

Abbreviations:

ADC

antibody-drug conjugate

ADCC

antibody-dependent cell-mediated cytotoxicity

ADR

adverse drug reaction

AGEP

acute generalized exanthematous pustulosis

AIHA

autoimmune hemolytic anemia

ARDS

acute respiratory distress syndrome

BAFF

B-cell activating factor

BAL

bronchoalveolar lavage

BOOP

bronchiolitis obliterans organizing pneumonia

CDC

complement-dependent cytotoxicity

CLL

chronic lymphoid leukemia

CRS

cytokine release syndrome

CTLA4

cytotoxic T lymphocyte-associated protein 4

DILD

drug-induced lung disease

DILI

drug-induced liver injury

DIV

drug-induced vasculitis

DRESS

drug reaction with eosinophilia and systemic symptoms

EGFR

epidermal growth factor receptor

EM

erythema multiforme

EMA

European Medicines Agency

EpCAM

epithelial cell adhesion molecule

FDA

Food and Drug Administration

FDE

fixed drug eruption

GCTB

giant cell tumor of the bone

GI

gastrointestinal

IR

infusion reaction

LON

late-onset neutropenia

LVD

left ventricular dysfunction

mAb

monoclonal antibody

MMAE

monomethyl auristatin E

NHL

non-Hodgkin's lymphoma

NSAID

non-steroidal anti-inflammatory drug

PML

progressive multifocal leukoencephalopathy

RANKL

receptor activator of nuclear factor κB ligand

SDF-1

stromal-derived factor-1

SIRS

systemic inflammatory response syndrome

SJS

Stevens-Johnson syndrome

TCR

T-cell receptor

TEN

toxic epidermal necrolysis

TLS

tumor lysis syndrome

TNFα

tumor necrosis factor α

VEGF

vascular endothelial growth factor

Footnotes

References

  • 1.World Health Organization International drug monitoring: the role of national centres. WHO Tec Rep Ser. 1972;498:1–25. [PubMed] [Google Scholar]
  • 2.Edwards IR, Aronson JK. Adverse drug reactions: definitions, diagnosis, and management. Lancet. 2000;356:1255–9. doi: 10.1016/S0140-6736(00)02799-9. [DOI] [PubMed] [Google Scholar]
  • 3.What is a serious event? Available at http:/www.fda.gov/safety/medwatch/howtoreport/ucm053087.htm (accessed February 18, 2013).
  • 4.Meyer UA. Pharmacogenetics and adverse drug reactions. Lancet. 2000;356:1667–71. doi: 10.1016/S0140-6736(00)03167-6. [DOI] [PubMed] [Google Scholar]
  • 5.Coombs RRA, Gell PGH. Classification of allergic reactions responsible for clinical hypersensitivity and disease. In: Gell PGH, Coombs RRA, Lachmann PJ, eds. Clinical Aspects of Immunology. Oxford: Blackwells, 1975: 761-81. [Google Scholar]
  • 6.Brockow K, Romano A, Aberer W, Bircher AJ, Barbaud A, Bonadonna P, Faria E, Kanny G, Lerch M, Pichler WJ, et al. European Network of Drug Allergy and the EAACI interest group on drug hypersensitivity Skin testing in patients with hypersensitivity reactions to iodinated contrast media - a European multicenter study. Allergy. 2009;64:234–41. doi: 10.1111/j.1398-9995.2008.01832.x. [DOI] [PubMed] [Google Scholar]
  • 7.Vane JR. The mode of action of aspirin and similar compounds. J Allergy Clin Immunol. 1976;58:691–712. doi: 10.1016/0091-6749(76)90181-0. [DOI] [PubMed] [Google Scholar]
  • 8.Baldo BA, Pham NH. Drug Allergy. Clinical Aspects, Diagnosis, Mechanisms, Structure-Activity Relationships. New York: Springer, 2013. [Google Scholar]
  • 9.Weiss RB, Baker JR., Jr. Hypersensitivity reactions from antineoplastic agents. Cancer Metastasis Rev. 1987;6:413–32. doi: 10.1007/BF00144273. [DOI] [PubMed] [Google Scholar]
  • 10.Ardavanis A, Tryfonopoulos D, Yiotis I, Gerasimidis G, Baziotis N, Rigatos G. Non-allergic nature of docetaxel-induced acute hypersensitivity reactions. Anticancer Drugs. 2004;15:581–5. doi: 10.1097/01.cad.0000131685.06390.b7. [DOI] [PubMed] [Google Scholar]
  • 11.Went P, Vasei M, Bubendorf L, Terracciano L, Tornillo L, Riede U, Kononen J, Simon R, Sauter G, Baeuerle PA. Frequent high-level expression of the immunotherapeutic target Ep-CAM in colon, stomach, prostate and lung cancers. Br J Cancer. 2006;94:128–35. doi: 10.1038/sj.bjc.6602924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Linke R, Klein A, Seimetz D. Catumaxomab: clinical development and future directions. MAbs. 2010;2:129–36. doi: 10.4161/mabs.2.2.11221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cragg MS, Walshe CA, Ivanov AO, Glennie MJ. The biology of CD20 and its potential as a target for mAb therapy. Curr Dir Autoimmun. 2005;8:140–74. doi: 10.1159/000082102. [DOI] [PubMed] [Google Scholar]
  • 14.Shim H. One target, different effects: a comparison of distinct therapeutic antibodies against the same targets. Exp Mol Med. 2011;43:539–49. doi: 10.3858/emm.2011.43.10.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jacene HA, Filice R, Kasecamp W, Wahl RL. Comparison of 90Y-ibritumomab tiuxetan and 131I-tositumomab in clinical practice. J Nucl Med. 2007;48:1767–76. doi: 10.2967/jnumed.107.043489. [DOI] [PubMed] [Google Scholar]
  • 16.Wahl RL. Tositumomab and (131)I therapy in non-Hodgkin’s lymphoma. J Nucl Med. 2005;46(Suppl 1):128S–40S. [PubMed] [Google Scholar]
  • 17.Jazirehi AR, Bonavida B. Cellular and molecular signal transduction pathways modulated by rituximab (rituxan, anti-CD20 mAb) in non-Hodgkin’s lymphoma: implications in chemosensitization and therapeutic intervention. Oncogene. 2005;24:2121–43. doi: 10.1038/sj.onc.1208349. [DOI] [PubMed] [Google Scholar]
  • 18.Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJT. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov. 2010;9:325–38. doi: 10.1038/nrd3003. [DOI] [PubMed] [Google Scholar]
  • 19.Zhang B. Ofatumumab. MAbs. 2009;1:326–31. doi: 10.4161/mabs.1.4.8895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cardarelli PM, Quinn M, Buckman D, Fang Y, Colcher D, King DJ, Bebbington C, Yarranton G. Binding to CD20 by anti-B1 antibody or F(ab’)(2) is sufficient for induction of apoptosis in B-cell lines. Cancer Immunol Immunother. 2002;51:15–24. doi: 10.1007/s00262-001-0247-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li S, Schmitz KR, Jeffrey PD, Wiltzius JJ, Kussie P, Ferguson KM. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell. 2005;7:301–11. doi: 10.1016/j.ccr.2005.03.003. [DOI] [PubMed] [Google Scholar]
  • 22.Freeman D, Sun J, Bass R, Jung K, Ogbagabriel S, Elliott G, et al Panitumumab and cetuximab epitope mapping and in vitro activity. J Clin Oncol. 2008;26(suppl 20) [Google Scholar]
  • 23.Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. doi: 10.1056/NEJMra043186. [DOI] [PubMed] [Google Scholar]
  • 24.Ghosh R, Narasanna A, Wang SE, Liu S, Chakrabarty A, Balko JM, González-Angulo AM, Mills GB, Penuel E, Winslow J, et al. Trastuzumab has preferential activity against breast cancers driven by HER2 homodimers. Cancer Res. 2011;71:1871–82. doi: 10.1158/0008-5472.CAN-10-1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Niculescu-Duvaz I. Trastuzumab emtansine, an antibody-drug conjugate for the treatment of HER2+ metastatic breast cancer. Curr Opin Mol Ther. 2010;12:350–60. [PubMed] [Google Scholar]
  • 26.Persson M, Tolmachev V, Andersson K, Gedda L, Sandström M, Carlsson J. [(177)Lu]pertuzumab: experimental studies on targeting of HER-2 positive tumour cells. Eur J Nucl Med Mol Imaging. 2005;32:1457–62. doi: 10.1007/s00259-005-1902-0. [DOI] [PubMed] [Google Scholar]
  • 27.Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM, Sliwkowski MX. Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell. 2004;5:317–28. doi: 10.1016/S1535-6108(04)00083-2. [DOI] [PubMed] [Google Scholar]
  • 28.Senter PD, Sievers EL. The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat Biotechnol. 2012;30:631–7. doi: 10.1038/nbt.2289. [DOI] [PubMed] [Google Scholar]
  • 29.Ellis LM. Mechanisms of action of bevacizumab as a component of therapy for metastatic colorectal cancer. Semin Oncol. 2006;33(Suppl 10):S1–7. doi: 10.1053/j.seminoncol.2006.08.002. [DOI] [PubMed] [Google Scholar]
  • 30.Alinari L, Lapalombella R, Andritsos L, Baiocchi RA, Lin TS, Byrd JC. Alemtuzumab (Campath-1H) in the treatment of chronic lymphocytic leukemia. Oncogene. 2007;26:3644–53. doi: 10.1038/sj.onc.1210380. [DOI] [PubMed] [Google Scholar]
  • 31.Hu Y, Turner MJ, Shields J, Gale MS, Hutto E, Roberts BL, Siders WM, Kaplan JM. Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology. 2009;128:260–70. doi: 10.1111/j.1365-2567.2009.03115.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hanley DA, Adachi JD, Bell A, Brown V. Denosumab: mechanism of action and clinical outcomes. Int J Clin Pract. 2012;66:1139–46. doi: 10.1111/ijcp.12022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Tarhini AA, Iqbal F. CTLA-4 blockade: therapeutic potential in cancer treatments. Onco Targets Ther. 2010;3:15–25. doi: 10.2147/OTT.S4833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.European Medicines Agency evaluation of medicines for human use. Assessment report for Removab. Proc. no. EMEA/H/C/000972, 2009. Available at http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-Public_assessment_report/human/000972/WC500051808.pdf (accessed at July 1, 2013).
  • 35.Therapeutic Goods Administration. Australian public assessment report for catumaxomab, October 2012. Available at http://www.tga.gov.au/pdf/auspar/auspar-catumaxomab-121004.pdf (accessed July 1, 2013).
  • 36.Zevalin® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125019s0156.pdf (accessed July 1, 2013).
  • 37.Bexxar® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/125011s102lbl.pdf (accessed July 1, 2013).
  • 38.Kaminski MS, Zelenetz AD, Press OW, Saleh M, Leonard J, Fehrenbacher L, Lister TA, Stagg RJ, Tidmarsh GF, Kroll S, et al. Pivotal study of iodine I 131 tositumomab for chemotherapy-refractory low-grade or transformed low-grade B-cell non-Hodgkin’s lymphomas. J Clin Oncol. 2001;19:3918–28. doi: 10.1200/JCO.2001.19.19.3918. [DOI] [PubMed] [Google Scholar]
  • 39.Adcetris® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/125388s0005lbl.pdf (accessed July 1, 2013).
  • 40.Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers EL, Forero-Torres A. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363:1812–21. doi: 10.1056/NEJMoa1002965. [DOI] [PubMed] [Google Scholar]
  • 41.Erbitux® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125084s168lbl.pdf (accessed July 1, 2013).
  • 42.Thomas M. Cetuximab: adverse event profile and recommendations for toxicity management. Clin J Oncol Nurs. 2005;9:332–8. doi: 10.1188/05.CJON.332-338. [DOI] [PubMed] [Google Scholar]
  • 43.Hoag JB, Azizi A, Doherty TJ, Lu J, Willis RE, Lund ME. Association of cetuximab with adverse pulmonary events in cancer patients: a comprehensive review. J Exp Clin Cancer Res. 2009;28:113. doi: 10.1186/1756-9966-28-113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Polito AJ. Pulmonary reactions to novel chemotherapeutic agents and biomolecules. In: Camus P, Rosenow III EC, eds. Drug-Induced and Iatrogenic Respiratory Disease. London: HodderArnold, 2010: 146-60. [Google Scholar]
  • 45.Rituxan® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/103705s5367s5388lbl.pdf (accessed July 1, 2013).
  • 46.Ram R, Bonstein L, Gafter-Gvili A, Ben-Bassat I, Shpilberg O, Raanani P. Rituximab-associated acute thrombocytopenia: an under-diagnosed phenomenon. Am J Hematol. 2009;84:247–50. doi: 10.1002/ajh.21372. [DOI] [PubMed] [Google Scholar]
  • 47.Wolach O, Bairey O, Lahav M. Late-onset neutropenia after rituximab treatment: case series and comprehensive review of the literature. Medicine (Baltimore) 2010;89:308–18. doi: 10.1097/MD.0b013e3181f2caef. [DOI] [PubMed] [Google Scholar]
  • 48.Le Guenno G, Ruivard M, Charra L, Philippe P. Rituximab-induced serum sickness in refractory immune thrombocytopenic purpura. Intern Med J. 2011;41:202–5. doi: 10.1111/j.1445-5994.2010.02384.x. [DOI] [PubMed] [Google Scholar]
  • 49.Dereure O, Navarro R, Rossi J-F, Guilhou J-J. Rituximab-induced vasculitis. Dermatology. 2001;203:83–4. doi: 10.1159/000051713. [DOI] [PubMed] [Google Scholar]
  • 50.Lioté H, Lioté F, Séroussi B, Mayaud C, Cadranel J. Rituximab-induced lung disease: A systematic literature review. Eur Respir J. 2010;35:681–7. doi: 10.1183/09031936.00080209. [DOI] [PubMed] [Google Scholar]
  • 51.Campath-1H® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2007/103948s5070lbl.pdf (accessed July 1, 2013).
  • 52.Lenihan DJ, Alencar AJ, Yang D, Kurzrock R, Keating MJ, Duvic M. Cardiac toxicity of alemtuzumab in patients with mycosis fungoides/Sézary syndrome. Blood. 2004;104:655–8. doi: 10.1182/blood-2003-07-2345. [DOI] [PubMed] [Google Scholar]
  • 53.Sun WL, Walder A, Fiegl M. Haemolytic anaemia triggered by antitumoural “biological agents” and immunotherapies with monoclonal antibodies: short review of the literature. memo. 2010;3:19–22. doi: 10.1007/s12254-010-0174-y. [DOI] [Google Scholar]
  • 54.Avastin® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/125085s0238lbl.pdf (accessed July 1, 2013).
  • 55.Shord SS, Bressler LR, Tierney LA, Cuellar S, George A. Understanding and managing the possible adverse effects associated with bevacizumab. Am J Health Syst Pharm. 2009;66:999–1013. doi: 10.2146/ajhp080455. [DOI] [PubMed] [Google Scholar]
  • 56.Sliesoraitis S, Tawfik B. Bevacizumab-induced bowel perforation. J Am Osteopath Assoc. 2011;111:437–41. [PubMed] [Google Scholar]
  • 57.Cortes J, Calvo V, Ramírez-Merino N, O’Shaughnessy J, Brufsky A, Robert N, Vidal M, Muñoz E, Perez J, Dawood S, et al. Adverse events risk associated with bevacizumab addition to breast cancer chemotherapy: a meta-analysis. Ann Oncol. 2012;23:1130–7. doi: 10.1093/annonc/mdr432. [DOI] [PubMed] [Google Scholar]
  • 58.Perjeta® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/125409lbl.pdf (accessed July 1, 2013).
  • 59.Hubalek M, Brantner C, Marth C. Role of pertuzumab in the treatment of HER2-positive breast cancer. Breast Cancer: Targets and Therapy. 2012;4:65–73. doi: 10.2147/BCTT.S23560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Herceptin® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5256lbl.pdf (accessed July 1, 2013).
  • 61.Parikh O, Neave F, Palmieri C. Severe thrombocytopenia induced by a single infusion of trastuzumab. Clin Breast Cancer. 2008;8:285–6. doi: 10.3816/CBC.2008.n.034. [DOI] [PubMed] [Google Scholar]
  • 62.Kadcyla® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf (accessed July 1, 2013).
  • 63.Xgeva® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125320s094lbl.pdf (accessed July 1, 2013).
  • 64.Yervoy® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/125377s0000lbl.pdf (accessed July 1, 2013).
  • 65.Weber J. Review: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse events. Oncologist. 2007;12:864–72. doi: 10.1634/theoncologist.12-7-864. [DOI] [PubMed] [Google Scholar]
  • 66.Tarhini A. Immune-mediated adverse events associated with ipilimumab CTLA-4 blockade therapy: The underlying mechanisms and clinical management. Scientifica. 2013 doi: 10.1155/2013/857519. Forthcoming. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Arzerra® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125326lbl.pdf (accessed July 1, 2013).
  • 68.Lemery SJ, Zhang J, Rothmann MD, Yang J, Earp J, Zhao H, McDougal A, Pilaro A, Chiang R, Gootenberg JE, et al. U.S. Food and Drug Administration approval: ofatumumab for the treatment of patients with chronic lymphocytic leukemia refractory to fludarabine and alemtuzumab. Clin Cancer Res. 2010;16:4331–8. doi: 10.1158/1078-0432.CCR-10-0570. [DOI] [PubMed] [Google Scholar]
  • 69.Vectibix® FDA: Full prescribing information. Adverse reactions. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125147s080lbl.pdf (accessed July 1, 2013).
  • 70.Stephens S, Emtage S, Vetterlein O, Chaplin L, Bebbington C, Nesbitt A, Sopwith M, Athwal D, Novak C, Bodmer M. Comprehensive pharmacokinetics of a humanized antibody and analysis of residual anti-idiotypic responses. Immunology. 1995;85:668–74. [PMC free article] [PubMed] [Google Scholar]
  • 71.Aster RH, Bougie DW. Drug-induced immune thrombocytopenia. N Engl J Med. 2007;357:580–7. doi: 10.1056/NEJMra066469. [DOI] [PubMed] [Google Scholar]
  • 72.Kenney B, Stack G. Drug-induced thrombocytopenia. Arch Pathol Lab Med. 2009;133:309–14. doi: 10.5858/133.2.309. [DOI] [PubMed] [Google Scholar]
  • 73.Salama A, Schütz B, Kiefel V, Breithaupt H, Mueller-Eckhardt C. Immune-mediated agranulocytosis related to drugs and their metabolites: mode of sensitization and heterogeneity of antibodies. Br J Haematol. 1989;72:127–32. doi: 10.1111/j.1365-2141.1989.tb07672.x. [DOI] [PubMed] [Google Scholar]
  • 74.Berliner N, Horwitz M, Loughran TP., Jr. Congenital and acquired neutropenia. Hematology Am Soc Hematol Educ Program. 2004:63–79. doi: 10.1182/asheducation-2004.1.63. [DOI] [PubMed] [Google Scholar]
  • 75.Garratty G. Drug-induced immune hemolytic anemia. Hematology Am Soc Hemat Educ Program 2009; 73-9. [DOI] [PubMed] [Google Scholar]
  • 76.Agache IO, Rogozea L. Management of hypersensitivity pneumonitis. Clin Translational Allergy. 2013;3:5. doi: 10.1186/2045-7022-3-5. Forthcoming. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Castell JV, Castell M. Allergic hepatitis induced by drugs. Curr Opin Allergy Clin Immunol. 2006;6:258–65. doi: 10.1097/01.all.0000235898.80052.80. [DOI] [PubMed] [Google Scholar]
  • 78.Adams DH, Ju C, Ramaiah SK, Uetrecht J, Jaeschke H. Mechanisms of immune-mediated liver injury. Toxicol Sci. 2010;115:307–21. doi: 10.1093/toxsci/kfq009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Apisamthanarax N, Duvic M. Dermatologic complications of cancer chemotherapy. In: Bast RC Jr, Kufe DW, Pollock RE, et al. eds. Holland-Frei Cancer Medicine, 5th edn. Hamilton, Ontario: BC Decker, 2000: 2271-8. [Google Scholar]
  • 80.Lynch TJ, Jr., Kim ES, Eaby B, Garey J, West DP, Lacouture ME. Epidermal growth factor receptor inhibitor-associated cutaneous toxicities: an evolving paradigm in clinical management. Oncologist. 2007;12:610–21. doi: 10.1634/theoncologist.12-5-610. [DOI] [PubMed] [Google Scholar]
  • 81.Balagula Y, Lacouture ME, Cotliar JA. Dermatologic toxicities of targeted anticancer therapies. J Support Oncol. 2010;8:149–61. [PubMed] [Google Scholar]
  • 82.Common terminology criteria for adverse events v3.0, DTCD, NCI, NIH, DHHS. Publish date: August 9, 2006 (http:/ctep.cancer.gov)
  • 83.Lenz H-J. Management and preparedness for infusion and hypersensitivity reactions. Oncologist. 2007;12:601–9. doi: 10.1634/theoncologist.12-5-601. [DOI] [PubMed] [Google Scholar]
  • 84.O'Neil BH. Overview of hypersensitivity to cancer therapies. Community Oncol. 2010;7:11–6. doi: 10.1016/S1548-5315(11)70383-7. [DOI] [Google Scholar]
  • 85.Joerger M. Prevention and handling of acute allergic and infusion reactions in oncology. Ann Oncol. 2012;23(Suppl 10):x313–9. doi: 10.1093/annonc/mds314. [DOI] [PubMed] [Google Scholar]
  • 86.Howard SC, Jones DP, Pui C-H. The tumor lysis syndrome. N Engl J Med. 2011;364:1844–54. doi: 10.1056/NEJMra0904569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bhatt V, Saleem A. Review: Drug-induced neutropenia--pathophysiology, clinical features, and management. Ann Clin Lab Sci. 2004;34:131–7. [PubMed] [Google Scholar]
  • 88.Lee A, ed. Adverse Drug Reactions, 2nd edn. London: Pharmaceutical Press, 2006. [Google Scholar]
  • 89.Uetrecht J, ed. Adverse Drug Reactions. Handbook of Experimental Pharmacology 196. Heidelberg: Springer-Verlag, 2010. [Google Scholar]
  • 90.Kimby E. Tolerability and safety of rituximab (MabThera) Cancer Treat Rev. 2005;31:456–73. doi: 10.1016/j.ctrv.2005.05.007. [DOI] [PubMed] [Google Scholar]
  • 91.Kang SP, Saif MW. Infusion-related and hypersensitivity reactions of monoclonal antibodies used to treat colorectal cancer--identification, prevention, and management. J Support Oncol. 2007;5:451–7. [PubMed] [Google Scholar]
  • 92.Chung CH, Mirakhur B, Chan E, Le QT, Berlin J, Morse M, Murphy BA, Satinover SM, Hosen J, Mauro D, et al. Cetuximab-induced anaphylaxis and IgE specific for galactose-α-1,3-galactose. N Engl J Med. 2008;358:1109–17. doi: 10.1056/NEJMoa074943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jankowitz R, Joyce J, Jacobs SA. Anaphylaxis after administration of ibritumomab tiuxetan for follicular non-hodgkin lymphoma. Clin Nucl Med. 2008;33:94–6. doi: 10.1097/RLU.0b013e31815ef825. [DOI] [PubMed] [Google Scholar]
  • 94.Vultaggio A, Matucci A, Nencini F, Pratesi S, Petroni G, Cammelli D, et al. Drug-specific Th2 cells and IgE antibodies in a patient with anaphylaxis to rituximab. In Arch Allergy Immunol 2012; 159:321-6. [DOI] [PubMed] [Google Scholar]
  • 95.Sallman DA, Macklin E, Ali S, Michaelson J, Barnes JA, Abramson JS, et al. Rituximab hypersensitivity reactions (HSR): Predictors of recurrent and severe reactions based on severity of reaction, NHL histologic subtype, and burden of disease. J Clin Oncol. 2012;30(Suppl) [Google Scholar]
  • 96.Dillman RO. Infusion reactions associated with the therapeutic use of monoclonal antibodies in the treatment of malignancy. Cancer Metastasis Rev. 1999;18:465–71. doi: 10.1023/A:1006341717398. [DOI] [PubMed] [Google Scholar]
  • 97.Chung CH. Managing premedications and the risk for reactions to infusional monoclonal antibody therapy. Oncologist. 2008;13:725–32. doi: 10.1634/theoncologist.2008-0012. [DOI] [PubMed] [Google Scholar]
  • 98.Winkler U, Jensen M, Manzke O, Schulz H, Diehl V, Engert A. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8) Blood. 1999;94:2217–24. [PubMed] [Google Scholar]
  • 99.Tonini G, Santini D, Vincenzi B, Borzomati D, Dicuonzo G, La Cesa A, Onori N, Coppola R. Oxaliplatin may induce cytokine-release syndrome in colorectal cancer patients. J Biol Regul Homeost Agents. 2002;16:105–9. [PubMed] [Google Scholar]
  • 100.Hayslip J, Fenning R. Safe administration of iodine-131 tositumomab after repeated infusion-related reactions to rituximab. Oncologist. 2007;12:338–40. doi: 10.1634/theoncologist.12-3-338. [DOI] [PubMed] [Google Scholar]
  • 101.Diehl LF, Ketchum LH. Autoimmune disease and chronic lymphocytic leukemia: autoimmune hemolytic anemia, pure red cell aplasia, and autoimmune thrombocytopenia. Semin Oncol. 1998;25:80–97. [PubMed] [Google Scholar]
  • 102.McLaughlin P, Grillo-López AJ, Link BK, Levy R, Czuczman MS, Williams ME, Heyman MR, Bence-Bruckler I, White CA, Cabanillas F, et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program. J Clin Oncol. 1998;16:2825–33. doi: 10.1200/JCO.1998.16.8.2825. [DOI] [PubMed] [Google Scholar]
  • 103.Cattaneo C, Spedini P, Casari S, Re A, Tucci A, Borlenghi E, Ungari M, Ruggeri G, Rossi G. Delayed-onset peripheral blood cytopenia after rituximab: frequency and risk factor assessment in a consecutive series of 77 treatments. Leuk Lymphoma. 2006;47:1013–7. doi: 10.1080/10428190500473113. [DOI] [PubMed] [Google Scholar]
  • 104.Otrock ZK, Mahfouz RA, Oghlakian GO, Salem ZM, Bazarbachi A. Rituximab-induced acute thrombocytopenia: a report of two cases. Haematologica. 2005;90(Suppl):ECR23. [PubMed] [Google Scholar]
  • 105.Cathomas R, Goldhirsch A, Von Moos R. Drug induced thrombocytopenia. N Engl J Med. 2007;357:1870–1. Correspondence. [PubMed] [Google Scholar]
  • 106.Jara Sánchez C, Olier Gárate C, García-Donas Jiménez J, Peñalver Párraga J. Drug-induced thrombocytopenia induced by trastuzumab: a special challenge in a curable disease. Ann Oncol. 2009;20:1607–8. doi: 10.1093/annonc/mdp374. [DOI] [PubMed] [Google Scholar]
  • 107.Haider I, Cahill M. Fatal thrombocytopaenia temporally related to the administration of alemtuzumab (MabCampath) for refractory CLL despite early discontinuation of therapy. Hematology. 2004;9:409–11. doi: 10.1080/10245330400001942. [DOI] [PubMed] [Google Scholar]
  • 108.Coles AJ, Compston DA, Selmaj KW, Lake SL, Moran S, Margolin DH, Norris K, Tandon PK, CAMMS223 Trial Investigators Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359:1786–801. doi: 10.1056/NEJMoa0802670. [DOI] [PubMed] [Google Scholar]
  • 109.Gibbs SD, Westerman DA, McCormack C, Seymour JF, Miles Prince H. Severe and prolonged myeloid haematopoietic toxicity with myelodysplastic features following alemtuzumab therapy in patients with peripheral T-cell lymphoproliferative disorders. Br J Haematol. 2005;130:87–91. doi: 10.1111/j.1365-2141.2005.05570.x. [DOI] [PubMed] [Google Scholar]
  • 110.Scappaticci FA, Skillings JR, Holden SN, Gerber HP, Miller K, Kabbinavar F, Bergsland E, Ngai J, Holmgren E, Wang J, et al. Arterial thromboembolic events in patients with metastatic carcinoma treated with chemotherapy and bevacizumab. J Natl Cancer Inst. 2007;99:1232–9. doi: 10.1093/jnci/djm086. [DOI] [PubMed] [Google Scholar]
  • 111.Nalluri SR, Chu D, Keresztes R, Zhu X, Wu S. Risk of venous thromboembolism with the angiogenesis inhibitor bevacizumab in cancer patients: a meta-analysis. JAMA. 2008;300:2277–85. doi: 10.1001/jama.2008.656. [DOI] [PubMed] [Google Scholar]
  • 112.Dunleavy K, Tay K, Wilson WH. Rituximab-associated neutropenia. Semin Hematol. 2010;47:180–6. doi: 10.1053/j.seminhematol.2010.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Voog E, Morschhauser F, Solal-Céligny P. Neutropenia in patients treated with rituximab. N Engl J Med. 2003;348:2691–4, discussion 2691-4. doi: 10.1056/NEJM200306263482620. [DOI] [PubMed] [Google Scholar]
  • 114.Papadaki T, Stamatopoulos K, Stavroyianni N, Paterakis G, Phisphis M, Stefanoudaki-Sofianatou K. Evidence for T-large granular lymphocyte-mediated neutropenia in Rituximab-treated lymphoma patients: report of two cases. Leuk Res. 2002;26:597–600. doi: 10.1016/S0145-2126(01)00183-7. [DOI] [PubMed] [Google Scholar]
  • 115.Liu JH, Wei S, Lamy T, Epling-Burnette PK, Starkebaum G, Djeu JY, Loughran TP. Chronic neutropenia mediated by fas ligand. Blood. 2000;95:3219–22. [PubMed] [Google Scholar]
  • 116.Papadaki T, Stamatopoulos K, Anagnostopoulos A, Fassas A. Rituximab-associated immune myelopathy. Blood. 2003;102:1557–8. doi: 10.1182/blood-2003-05-1532. [DOI] [PubMed] [Google Scholar]
  • 117.Terrier B, Ittah M, Tourneur L, Louache F, Soumelis V, Lavie F, Casadevall N, Candon S, Hummel A, Mariette X, et al. Late-onset neutropenia following rituximab results from a hematopoietic lineage competition due to an excessive BAFF-induced B-cell recovery. Haematologica. 2007;92:e20–3. doi: 10.3324/haematol.11031. [DOI] [PubMed] [Google Scholar]
  • 118.Jourdan E, Topart D, Richard B, Jourdan J, Sotto A. Severe autoimmune hemolytic anemia following rituximab therapy in a patient with a lymphoproliferative disorder. Leuk Lymphoma. 2003;44:889–90. doi: 10.1080/1042819021000055110. [DOI] [PubMed] [Google Scholar]
  • 119.Ramamoorthy SK, Marangolo M, Durrant E, Akima S, Gottlieb DJ. Unusual reaction to Rituximab with intra-vascular hemolysis, rhabdomyolysis, renal failure and bone marrow necrosis. Leuk Lymphoma. 2006;47:747–50. doi: 10.1080/10428190500399193. [DOI] [PubMed] [Google Scholar]
  • 120.Ruch J, McMahon B, Ramsey G, Kwaan HC. Catastrophic multiple organ ischemia due to an anti-Pr cold agglutinin developing in a patient with mixed cryoglobulinemia after treatment with rituximab. Am J Hematol. 2009;84:120–2. doi: 10.1002/ajh.21330. [DOI] [PubMed] [Google Scholar]
  • 121.Elimelakh M, Dayton V, Park KS, Gruessner AC, Sutherland D, Howe RB, Reding MT, Eastlund T, van Burik JA, Singleton TP, et al. Red cell aplasia and autoimmune hemolytic anemia following immunosuppression with alemtuzumab, mycophenolate, and daclizumab in pancreas transplant recipients. Haematologica. 2007;92:1029–36. doi: 10.3324/haematol.10733. [DOI] [PubMed] [Google Scholar]
  • 122.Kako S, Kanda Y, Oshima K, Nishimoto N, Sato H, Watanabe T, Hosoya N, Motokura T, Miyakoshi S, Taniguchi S, et al. Late onset of autoimmune hemolytic anemia and pure red cell aplasia after allogeneic hematopoietic stem cell transplantation using in vivo alemtuzumab. Am J Hematol. 2008;83:247–9. doi: 10.1002/ajh.21086. [DOI] [PubMed] [Google Scholar]
  • 123.Fiegl M, Falkner A, Hopfinger G, Brugger S, Zabernigg A, Bauer F, et al. Routine clinical use of alemtuzumab in patients with heavily pretreated B-cell chronic lymphocytic leukemia: a nation-wide retrospective study in Austria. Cancer. 2006;107:2408–16. doi: 10.1002/cncr.22263. [DOI] [PubMed] [Google Scholar]
  • 124.Wierda WG, Kipps TJ, Keating MJ, Brown JR, Gribben JG, Browning M, Rassenti LZ, Greaves AW, Neuberg D, O’Brien SM, CLL Research Consortium Self-administered, subcutaneous alemtuzumab to treat residual disease in patients with chronic lymphocytic leukemia. Cancer. 2011;117:116–24. doi: 10.1002/cncr.25379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Frangié C, Lefaucheur C, Medioni J, Jacquot C, Hill GS, Nochy D. Renal thrombotic microangiopathy caused by anti-VEGF-antibody treatment for metastatic renal-cell carcinoma. Lancet Oncol. 2007;8:177–8. doi: 10.1016/S1470-2045(07)70037-2. [DOI] [PubMed] [Google Scholar]
  • 126.Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, et al. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med. 2008;358:1129–36. doi: 10.1056/NEJMoa0707330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Anandacoomarasamy A, Kannangara S, Barnsley L. Cutaneous vasculitis associated with infliximab in the treatment of rheumatoid arthritis. Intern Med J. 2005;35:638–40. doi: 10.1111/j.1445-5994.2005.00899.x. [DOI] [PubMed] [Google Scholar]
  • 128.Kandula P, Kouides PA. Rituximab-induced leukocytoclastic vasculitis: a case report. Arch Dermatol. 2006;142:243–53. doi: 10.1001/archderm.142.2.246. [DOI] [PubMed] [Google Scholar]
  • 129.Kim MJ, Kim HO, Kim HY, Park YM. Rituximab-induced vasculitis: A case report and review of the medical published work. J Dermatol. 2009;36:284–7. doi: 10.1111/j.1346-8138.2009.00639.x. [DOI] [PubMed] [Google Scholar]
  • 130.D’Arcy CA, Mannik M. Serum sickness secondary to treatment with the murine-human chimeric antibody IDEC-C2B8 (rituximab) Arthritis Rheum. 2001;44:1717–8. doi: 10.1002/1529-0131(200107)44:7&#x0003c;1717::AID-ART299&#x0003e;3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 131.Hellerstedt B, Ahmed A. Delayed-type hypersensitivity reaction or serum sickness after rituximab treatment. Ann Oncol. 2003;14:1792. doi: 10.1093/annonc/mdg488. [DOI] [PubMed] [Google Scholar]
  • 132.Finger E, Scheinberg M. Development of serum sickness-like symptoms after rituximab infusion in two patients with severe hypergammaglobulinemia. J Clin Rheumatol. 2007;13:94–5. doi: 10.1097/01.rhu.0000262585.18582.1e. [DOI] [PubMed] [Google Scholar]
  • 133.Fong L, Small EJ. Anti-cytotoxic T-lymphocyte antigen-4 antibody: the first in an emerging class of immunomodulatory antibodies for cancer treatment. J Clin Oncol. 2008;26:5275–83. doi: 10.1200/JCO.2008.17.8954. [DOI] [PubMed] [Google Scholar]
  • 134.Maker AV, Attia P, Rosenberg SA. Analysis of the cellular mechanism of antitumor responses and autoimmunity in patients treated with CTLA-4 blockade. J Immunol. 2005;175:7746–54. doi: 10.4049/jimmunol.175.11.7746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Weber J. Review: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse events. Oncologist. 2007;12:864–72. doi: 10.1634/theoncologist.12-7-864. [DOI] [PubMed] [Google Scholar]
  • 136.Agache IO, Rogozea L. Management of hypersensitivity pneumonitis. Clin Translational Allergy. 2013;3:5. doi: 10.1186/2045-7022-3-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ergin AB, Fong N, Daw HA. Rituximab-induced bronchiolitis obliterans organizing pneumonia. Case Rep Med. 2012;2012:680431. doi: 10.1155/2012/680431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Burton C, Kaczmarski R, Jan-Mohamed R. Interstitial pneumonitis related to rituximab therapy. N Engl J Med. 2003;348:2690–1, discussion 2690-1. doi: 10.1056/NEJM200306263482619. [DOI] [PubMed] [Google Scholar]
  • 139.Wagner SA, Mehta AC, Laber DA. Rituximab-induced interstitial lung disease. Am J Hematol. 2007;82:916–9. doi: 10.1002/ajh.20910. [DOI] [PubMed] [Google Scholar]
  • 140.Kishi J, Nanki T, Watanabe K, Takamura A, Miyasaka N. A case of rituximab-induced interstitial pneumonitis observed in systemic lupus erythematosus. Rheumatology (Oxford) 2009;48:447–8. doi: 10.1093/rheumatology/kep015. [DOI] [PubMed] [Google Scholar]
  • 141.Hadjinicolaou AV, Nisar MK, Parfrey H, Chilvers ER, Ostör AJK. Non-infectious pulmonary toxicity of rituximab: a systematic review. Rheumatology (Oxford) 2012;51:653–62. doi: 10.1093/rheumatology/ker290. [DOI] [PubMed] [Google Scholar]
  • 142.Chaumais MC, Garnier A, Chalard F, Peuchmaur M, Dauger S, Jacqz-Agrain E, Deschênes G. Fatal pulmonary fibrosis after rituximab administration. Pediatr Nephrol. 2009;24:1753–5. doi: 10.1007/s00467-009-1195-9. [DOI] [PubMed] [Google Scholar]
  • 143.Kamei K, Ito S, Iijima K. Severe respiratory adverse events associated with rituximab infusion. Pediatr Nephrol. 2010;25:1193. doi: 10.1007/s00467-009-1408-2. [DOI] [PubMed] [Google Scholar]
  • 144.Child N, O’Carroll M, Berkahn L. Rituximab-induced interstitial lung disease in a patient with immune thrombocytopenia purpura. Intern Med J. 2012;42:e12–4. doi: 10.1111/j.1445-5994.2011.02701.x. [DOI] [PubMed] [Google Scholar]
  • 145.Arulkumaran N, Suleman R, Cecconi M, Kiely P, Chua F. Rituximab associated pneumonitis in antineutrophil cytoplasmic antibody-associated vasculitis. J Clin Rheumatol. 2012;18:39–41. doi: 10.1097/RHU.0b013e31823ee5bf. [DOI] [PubMed] [Google Scholar]
  • 146.Montero AJ, McCarthy JJ, Chen G, Rice L. Acute respiratory distress syndrome after rituximab infusion. Int J Hematol. 2005;82:324–6. doi: 10.1532/IJH97.NA0506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Benyunes MC, Multani PS, Saunders A. Interstitial pneumonitis related to rituximab therapy. N Engl J Med. 2003;348:2691. doi: 10.1056/NEJM200306263482619. [DOI] [PubMed] [Google Scholar]
  • 148.Alho HS, Maasilta PK, Harjula AL, Hämmäinen P, Salminen J, Salminen US. Tumor necrosis factor-alpha in a porcine bronchial model of obliterative bronchiolitis. Transplantation. 2003;76:516–23. doi: 10.1097/01.TP.0000074700.30536.76. [DOI] [PubMed] [Google Scholar]
  • 149.Alexandrescu DT, Dutcher JP, O’Boyle K, Albulak M, Oiseth S, Wiernik PH. Fatal intra-alveolar hemorrhage after rituximab in a patient with non-Hodgkin lymphoma. Leuk Lymphoma. 2004;45:2321–5. doi: 10.1080/10428190410001697359. [DOI] [PubMed] [Google Scholar]
  • 150.Gonҫalo M, Bruynzeel DP. Mechanisms in cutaneous drug hypersensitivity reactions. In: Zhai H, Wilhelm K-P, Maibach HI, eds. Marzulli and Maibach’s Dermatotoxicology, 7th edn. Boca Raton Florida: CRC Press, 2008: 259-68. [Google Scholar]
  • 151.Lowndes S, Darby A, Mead G, Lister A. Stevens-Johnson syndrome after treatment with rituximab. Ann Oncol. 2002;13:1948–50. doi: 10.1093/annonc/mdf350. [DOI] [PubMed] [Google Scholar]
  • 152.Scheinfeld N. A review of rituximab in cutaneous medicine. Dermatol Online J. 2006;12:3. [PubMed] [Google Scholar]
  • 153.Segaert S, Van Cutsem E. Clinical signs, pathophysiology and management of skin toxicity during therapy with epidermal growth factor receptor inhibitors. Ann Oncol. 2005;16:1425–33. doi: 10.1093/annonc/mdi279. [DOI] [PubMed] [Google Scholar]
  • 154.Melosky B, Burkes R, Rayson D, Alcindor T, Shear N, Lacouture M. Management of skin rash during EGFR-targeted monoclonal antibody treatment for gastrointestinal malignancies: Canadian recommendations. Curr Oncol. 2009;16:16–26. doi: 10.3747/co.v16i1.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Mitchell EP, Pérez-Soler R, Van Cutsem E, Lacouture ME. Clinical presentation and pathophysiology of EGFRI dermatologic toxicities. Oncology (Williston Park) 2007;21(Suppl 5):4–9. [PubMed] [Google Scholar]
  • 156.Klein PM, Dybdal N. Trastuzumab and cardiac dysfunction: update on preclinical studies. Semin Oncol. 2003;30(Suppl 16):49–53. doi: 10.1053/j.seminoncol.2003.08.007. [DOI] [PubMed] [Google Scholar]
  • 157.Crone SA, Zhao YY, Fan L, Gu Y, Minamisawa S, Liu Y, Peterson KL, Chen J, Kahn R, Condorelli G, et al. ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat Med. 2002;8:459–65. doi: 10.1038/nm0502-459. [DOI] [PubMed] [Google Scholar]
  • 158.Grazette LP, Boecker W, Matsui T, Semigran M, Force TL, Hajjar RJ, Rosenzweig A. Inhibition of ErbB2 causes mitochondrial dysfunction in cardiomyocytes: implications for herceptin-induced cardiomyopathy. J Am Coll Cardiol. 2004;44:2231–8. doi: 10.1016/j.jacc.2004.08.066. [DOI] [PubMed] [Google Scholar]
  • 159.Howard SC, Jones DP, Pui C-H. The tumor lysis syndrome. N Engl J Med. 2011;364:1844–54. doi: 10.1056/NEJMra0904569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Brooks B. Progressive multifocal leukoencephalopathy. In: Gilman S ed. Neurobiology of Disease. London: Elsevier Academic Press, 2007: 185-96. [Google Scholar]
  • 161.Carson KR, Evens AM, Richey EA, Habermann TM, Focosi D, Seymour JF, Laubach J, Bawn SD, Gordon LI, Winter JN, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood. 2009;113:4834–40. doi: 10.1182/blood-2008-10-186999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Johansson SGO, Bieber T, Dahl R, Friedmann PS, Lanier BQ, Lockey RF, Motala C, Ortega Martell JA, Platts-Mills TA, Ring J, et al. Revised nomenclature for allergy for global use: Report of the Nomenclature Review Committee of the World Allergy Organization, October 2003. J Allergy Clin Immunol. 2004;113:832–6. doi: 10.1016/j.jaci.2003.12.591. [DOI] [PubMed] [Google Scholar]
  • 163.Reese JA, Li X, Hauben M, Aster RH, Bougie DW, Curtis BR, George JN, Vesely SK. Identifying drugs that cause acute thrombocytopenia: an analysis using 3 distinct methods. Blood. 2010;116:2127–33. doi: 10.1182/blood-2010-03-276691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Arnold DM, Santoso S, Greinacher A, Platelet Immunology Scientific Subcommittee of the ISTH Recommendations for the implementation of platelet autoantibody testing in clinical trials of immune thrombocytopenia. J Thromb Haemost. 2012;10:695–7. doi: 10.1111/j.1538-7836.2012.04664.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Stroncek DF. Drug-induced immune neutropenia. Transfus Med Rev. 1993;7:268–74. doi: 10.1016/S0887-7963(93)70146-7. [DOI] [PubMed] [Google Scholar]
  • 166.Radić M, Martinović Kaliterna D, Radić J. Drug-induced vasculitis: a clinical and pathological review. Neth J Med. 2012;70:12–7. [PubMed] [Google Scholar]
  • 167.Matsuno O. Drug-induced interstitial lung disease: mechanisms and best diagnostic approaches. Respir Res. 2012;13:39. doi: 10.1186/1465-9921-13-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ellis SJ, Cleverley JR, Müller NL. Drug-induced lung disease: high-resolution CT findings. AJR Am J Roentgenol. 2000;175:1019–24. doi: 10.2214/ajr.175.4.1751019. [DOI] [PubMed] [Google Scholar]
  • 169.Epler GR. Bronchiolitis obliterans organizing pneumonia, 25 years: a variety of causes, but what are the treatment options? Expert Rev Respir Med. 2011;5:353–61. doi: 10.1586/ers.11.19. [DOI] [PubMed] [Google Scholar]

Articles from Oncoimmunology are provided here courtesy of Taylor & Francis

RESOURCES