Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2019 Nov 22:251–348. doi: 10.1016/B978-0-323-67509-3.00016-0

Passive Monoclonal and Polyclonal Antibody Therapies

J Peter R Pelletier 1, Faisal Mukhtar 1
Editor: Robert W Maitta1
PMCID: PMC7153350

Abstract

Passive antibody therapies have a long history of use. In the 19th century, antibodies from xenographic sources of polyclonal antibodies were used to treat infections (diphtheria). They were used often as protection from infectious agents and toxins. Complications related to their use involved development of immune complexes and severe allergic reactions. As a result, human source plasma for polyclonal antibodies became the preferential source for antibodies. They are used to treat infection, remove toxins, prevent hemolytic disease of the newborn, modify inflammatory reactions, and control autoimmune diseases. Continued improvements in processing decreased the transfusion/infusion transmission of infections. In the late 20th century (∼1986), monoclonal antibodies were developed. The first monoclonal antibodies were of xenographic source and were wrought with problems of immunogenicity. These forms of antibodies did not gain favor until chimerization took pace in the mid-1990s and in 1998 two monoclonal antibodies were approved one to treat respiratory syncytial virus and the other for breast cancers. Further development of humanized and then fully human monoclonal antibodies has led to an evolution of therapies with these agents. Monoclonal antibodies are being researched or approved to treat a multitude of diseases to include oncologic, inflammatory, autoimmune, cardiovascular, respiratory, neurologic, allergic, benign hematologic, infections, orthopedic, coagulopathy, metabolic and to decrease morbidity of disease (diminution of pain), modify disease progression, and potentially anatomic development. In this chapter, we will review the history of use of these passive antibody therapies, their mechanism of action, pharmacologic-therapeutic classification, particular medical indication, adverse reactions, and potential future use of these medications.

Keywords: Antibodies, Bispecific T-cell engager, Chimeric, Humanized, Monoclonal, Polyclonal, Trimeric

Passive Polyclonal Antibodies Therapy

Passive Polyclonal Antibody Treatment Overview

Polyclonal immunoglobulins have been in use since the 19th century to protect against infectious agents, toxins, and disease conditions such as those with an autoimmune etiology. These immunoglobulin preparations are made from pools of selected human donors or animals with high titers of antibodies against viruses and toxins. These antibody treatments provide passive transfer of high titer antibodies that either reduces risk or reduces severity of infection. They are used to prevent hemolytic disease of the newborn and modify inflammatory reactions. Earlier drugs were very nonselective and patients frequently succumbed to infection due to suppression of both antibody-mediated (humoral) and cell-mediated arms of the immune system. Today, the principal approach is to alter lymphocyte function using drugs or antibodies against immune proteins. However, with the advent of human organ and tissue transplantation (e.g., kidney, heart, bone marrow, and/or peripheral blood stem cells) as treatment options, these polyclonal antibody therapies in combination with other treatment regimens are being used to lower the ability of the body's immune system to reject these transplants. However, their use is not without risk, as complications include development of immune complexes and severe allergic reactions. A summary of these polyclonal antibody therapies may be found in Table 16.2.

Table 16.2.

Summary of Polyclonal Antibody Therapies.

Generic Drug Name Brand Name Additional Brand Names AHFS Classification Dosage Form(s) Restricted Medication
Antithymocyte globulin (equine) Atgam Immunosuppressive agent Intravenous solution
Antithymocyte globulin (rabbit) Thymoglobulin Immunosuppressive agent Intravenous solution
Antivenin Latrodectus mactans Black widow
Antivenin
Serums Intravenous solution
Antivenin micrurus Eastern and
Texas coral
Snake
Antivenin
Serums Intravenous solution
Botulism immune globulin BabyBIG
Crotalidae polyvalent immune Fab Crofab Serums Intravenous solution
Cytomegalovirus immune globulin Cytogam Serums Intravenous solution Yes
Digoxin immune Fab Digibind Serums Intravenous solution
Hepatitis B immune globulin Hepagam-B Serums Intramuscular solution,
Intravenous solution
Hepatitis B immune globulin BayHepB HepaGam B, Hyper Hep B, Nabi-HB
High antibody titer Ebola FFP
High antibody titer influenza FFP
Immunoglobulin (generic) Gamunex Vivaglobin, Cuvitru, Privigen, gammagard, octagam, gamunex, hizentra, Bivigam, Carimune, Flebogamma, Gamastan, Gamimune, Gammaplex, gammar, Panglobulin, Panzyga, Sandoglobulin Intravenous,
Subcutaneous
Treat XLA, CVID, Hyper IgM syndromes, Wiskott Aldrich syndrome
Rabies immune globulin Bayrab HyperRAB, Imogam rabies, KedRAB
Respiratory syncytial virus immune globulin RespiGam
Rho (D) immune globulin WhinRho
RhoGam
Rhophylac, MicRhoGAM, BatRhoD, HyperRho Serums Intravenous, intramuscular solutions
Rimabotulinumtoxin B Myobloc Other Miscellaneous
Therapeutic agents
Injection solution Yes
Rozrolimupab Anti-RhD
Prevent isoimmunization
ITP
Tetanus immune globulin Baytet Hypertet
Varicella zoster immune globulin VariZIG

Immunosuppressive Agents: Disease Modifying

Antithymocyte globulin (rabbit)/thymoglobulin; antithymocyte globulin (equine)/Atgam

Description

Rabbit antithymocyte globulin (rATG) and equine antithymocyte globulin (eATG) are purified, pasteurized preparation of lymphocyte depleting polyclonal gamma immunoglobulin (IgG) raised against human thymus lymphocytes in rabbits and horses, respectively. They are used in prevention and/or treatment of renal transplant rejection worldwide.1, 2, 3, 4, 5, 6, 7

History of antibody use

rATG induction in combination with immunosuppressive therapy is more effective in preventing episodes of acute renal graft rejection in adult renal transplant recipients, in recurrent episodes of acute rejection,8, 9 and those acute rejections that are not responsive to high-dose corticosteroid therapy than other monoclonal antibody preparations.10, 11 rATG recipients had a lower incidence of biopsy-confirmed acute rejection episodes,12 greater event-free survival up to 10 years posttransplantation, and greater graft survival up to 5 years posttransplantation.13

Mechanisms of action

The exact mechanism of these polyclonal antibodies has not been fully understood.3, 4, 14, 15, 16, 17, 18, 19, 20 However, being polyclonal, they display specificity toward a wide variety of surface antigens (Ags) expressed on T and B-lymphocytes, dendritic cells, natural killer (NK) cells, and endothelial cells. However, T-cell depletion is considered to play a key role by modulating the expression of lymphocyte surface antigens involved in a wide variety of functions such as T-cell activation to endothelial adherence, activation of certain transcription factors, and interference with numerous immune cell processes, such as cytokine production, chemotaxis, endocytosis, cell stimulation, and proliferation.14, 15, 16, 17, 18, 19, 20

In vitro studies indicate that binding of eATG to cells is generally nonspecific; the drug binds to visceral tissues, including thymus and testis cell membranes and nuclear and cytoplasmic components of tissues such as tonsil, kidney, and liver,21 and is extensively bound to bone marrow cells,22 and to other peripheral blood cells besides lymphocytes.21

Diseases treated

As mentioned earlier, both antithymocyte globulins are used for treatment and prevention of acute renal allograft rejection.2, 3, 4, 5, 6, 7, 8 More rATG recipients have been reported to achieve the endpoint of successful response (return of serum creatinine levels to baseline by end of treatment or within 14 days of treatment initiation). However, among those who achieved a successful response, fewer episodes of recurrent rejection occurred with rATG within 90 days of treatment cessation.2 eATG is also used for treating moderate-to-severe aplastic anemia in patients who are unsuitable for bone marrow transplantation.3, 23, 24

Adverse effects

The most common adverse effects are fever, thrombocytopenia, leukopenia, gastrointestinal disorders, and/or concurrent infection.1, 2 Cytomegalovirus (CMV) infection was generally higher with rATG except in high-risk patients.1, 25 eATG therapy may result in reactivation of or infection with CMV, herpes simplex virus,25 or Epstein–Barr virus.26 The incidence of malignancies is generally lower with rATG therapy.27 This product is made of equine and human blood components, so it may carry a risk of transmitting infectious agents such as viruses, and theoretically, the Creutzfeldt–Jakob disease (CJD) agent.

Update

There has been recent evidence that the addition of human anti-T-lymphocyte globulin (ATLG) plus cyclosporine and methotrexate to standard graft-versus-host disease (GVHD) prophylaxis is preferred over standard GVHD prophylaxis alone because it improves the probability of survival without relapse and of chronic GVHD after myeloablative peripheral blood stem-cell transplantation from a human leukocyte antigen (HLA)-identical sibling donor for patients with acute leukemia in remission. Additionally, this therapy provides better quality of life and shorter immunosuppressive treatment compared to standard GVHD prophylaxis without ATLG.22

Antitoxin and Immune Globulins: Disease Modifying

Tetanus immune globulin/Baytet/Hypertet

Description

Tetanus immune globulin (TIG) is a specific solvent-detergent-treated plasma-derived product obtained from donors immunized with tetanus toxoid. TIG contains tetanus antitoxin that provides temporary passive immunity to individuals who have low or no immunity to the toxin produced by Clostridium tetani.28, 29

Mechanisms of action

TIG contains tetanus antitoxin antibodies, which neutralize the free form of the powerful exotoxin produced by Clostridium tetani.28, 30 TIG can only neutralize unbound exotoxin; it does not affect toxin already bound to nerve endings.31

Diseases treated

TIG is used to provide passive immunity to tetanus as part of a postexposure prophylaxis regimen following an injury in patients whose immunization is incomplete or uncertain or if it has been more than 10 years since last dose of tetanus toxoid.1, 3, 4, 5, 6, 7, 8, 9

Adverse reaction

Slight soreness at injection site, mild fever, and rarely sensitization to repeated injections of human immune globulin has been reported.28

Antitoxin and Immune Globulins: Disease Modifying

Cytomegalovirus immune Globulin/Cytogam

Description

Cytomegalovirus immune globulin IV (CMV-IG) is a purified immune globulin (hyperimmune globulin) that contains immunoglobulin G (IgG) derived from pooled adult human plasma selected for high titers of anti-CMV antibodies.32

Mechanisms of action

CMV-IG provides relatively high concentration of antibodies directed against CMV. It provides prophylaxis against CMV infection or disease in immunocompromised individuals.32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43 Results from in vitro studies and mice indicate that anti-CMV antibodies can neutralize the pathogenic properties of CMV.42, 43, 44 As CMV usually targets a population of bone marrow-derived myeloid lineage progenitor cells, antibody-neutralization of the virus alone may not be enough to prevent or make active disease less severe in already CMV-infected individuals.42, 44, 45, 46, 47

Disease treated

CMV-IG provides passive immunity to individuals who are at risk for primary CMV infection/disease, or secondary CMV disease (reactivation of CMV).41, 42, 44, 45, 46, 48, 49, 50, 51 It is also prescribed for the prophylaxis of CMV disease associated with transplantation of kidney, lung, liver, pancreas, and heart. With the exception of CMV-seronegative recipients of kidneys from CMV-seropositive donors, CMV-IG prophylaxis should be considered in conjunction with ganciclovir.

Adverse reactions

Most frequent adverse reactions reported are flushing, chills, muscle cramps, back pain, fever, nausea, vomiting, arthralgia, and wheezing.32, 34, 35, 36 There is a slight risk of hemolysis, as intravenous immunoglobulin (IVIG) products can contain blood group antibodies, which may act as a hemolysin and induce in vivo coating of red blood cells with immunoglobulin, causing a positive direct antiglobulin reaction. Transfusion-related acute lung injury (noncardiogenic pulmonary edema) and thrombotic events have been reported in patients receiving IVIG preparations.32

Similar to all other products made from human plasma, this CMV-IG also carries the possibility for transmission of blood-borne viral agents and the CJD agent. However, this IVIG is treated with a solvent detergent viral inactivation procedure to inactivate a wide spectrum of lipid-enveloped viruses, including HIV-1, HIV-2, Hepatitis B, and Hepatitis C.

Antivenin [latrodectus mactans]/black widow spider antivenin—antivenin Micrurus fulvius/eastern and Texas coral snake antivenin—crotalidae polyvalent immune Fab/Crofab

Description

These antivenins are sterile, nonpyrogenic, purified, and lyophilized preparation of specific venom-neutralizing serum globulins obtained from the blood serum of healthy horses exposed to the venom of black widow spiders and eastern coral snake (Micrurus fulvius) venom, respectively.52, 53, 54, 55 In contrast, crofab is an antivenin made up of ovine Fab (monovalent) immunoglobulin fragments obtained from blood of healthy sheep immunized with North American Crotalinae subfamily of venomous snakes that includes rattlesnakes, copperheads, cottonmouth, or water moccasins.56

Mechanisms of action

Mode of action of these antivenins is unknown.52 However, they probably act by neutralizing venom of black widow spiders and coral snakes.54 Crofab is a venom-specific Fab fragment of IgG that works by binding and neutralizing venom toxins, facilitating their redistribution away from target tissues and their elimination from the body.56

Disease treated

These antivenins are indicated for patients with symptoms due to bites by black widow spider (Latrodectus mactans)52 and bites of two genera of coral snakes, that is, Micrurus (including the eastern and Texas varieties) and Micruroides (the Sonoran or Arizona variety), found in southeastern Arizona and southwestern New Mexico.52, 57, 58, 59 Antivenin Micrurus fulvius (equine origin) is indicated only for treatment and management of adult and pediatric patients exposed to North American crotalid envenomation.54

Adverse effects

Immediate systemic reactions (allergic reactions or anaphylaxis) and death can occur in patients sensitive to antivenin from horse serum.52, 60 Most common adverse reactions to crofab are urticaria, rash, nausea, pruritus, and back pain.61, 62

High antibody titer influenza fresh frozen plasma

Description

Use of convalescent (persons who have recovered from a particular infection) donor plasma with high hemagglutination inhibition titer against certain influenza strains has been recommended as a primary therapy for severe respiratory infectious diseases including influenza, severe acute respiratory syndrome, and Middle East respiratory syndrome.63

History of antibody use

A meta-analysis of previous cohort studies during the 1918 influenza pandemic showed a case-fatality rate of 16% among subjects treated with plasma, serum, or whole blood compared to 37% among controls. Similarly, in 2009, a cohort study using convalescent plasma for the treatment of pandemic H1N1 influenza resulted in a mortality of 20% in the treatment group versus 54% in the control group.64

Mechanisms of action

Antiinfluenza convalescent plasma decreases the rate of viral shedding measured by neutralizing antibody titer and hemagglutination inhibition.65 Both preexisting immunity (previous infections and vaccinations) as well as any immune response occurring after illness onset makes this mechanism of action more complex.

Disease classifications treated

Influenza, severe acute respiratory syndrome, and Middle East respiratory syndrome.63

Adverse effects

Convalescent plasma seems safe. The serious adverse events reported are related to the underlying influenza, its complications, preexisting comorbidities, and not due to the convalescent plasma usage.

High antibody titer ebola fresh frozen plasma

Description

Antibodies to the Ebola virus (EV) in whole blood or plasma from convalescent donors may be effective in the treatment of EV infection.

History of antibody use

The World Health Organization (WHO) has stated that convalescent blood or plasma is an option in the treatment of Ebola.66 In 1999, transfusion of locally collected convalescent blood helped to decrease Ebola mortality.67 Therefore, WHO has recommended the collection of convalescent plasma to treat patients with Ebola virus infection.

Mechanisms of action

This fresh frozen plasma (FFP) has high titers of antibodies directed against Ebola virus.68

Adverse effects

Convalescent plasma seems safe with few adverse effects.69, 70

Digoxin immune Fab/DigiFab; Digibind

Description

Digoxin immune Fab is a sterile, purified, lyophilized monovalent preparation of bovine immunoglobulin Fab fragments that binds to digoxin. These Fab fragments are obtained from the blood of healthy sheep immunized with a digoxin derivative, digoxindicarboxymethoxylamine, a digoxin analogue that contains the functionally essential cyclopentaperhydrophenanthrene: lactone ring moiety coupled to keyhole limpet hemocyanin. The final product is prepared by taking the immunoglobulin fraction of the ovine serum, digesting it with papain, and isolating the digoxin-specific Fab fragments by affinity chromatography.71, 72, 73, 74, 75, 76, 77, 78, 79

Mechanisms of action

DigiFab or Digibind have antigen-binding fragments that bind to free digoxin molecules that results in an equilibrium shift away from binding to receptors, thereby reversing the cardiotoxic effects of the glycoside.71, 72, 75, 76, 78, 80, 81, 82, 83, 84, 85, 86, 87 Subsequently, Fab-digoxin complexes are cleared by the kidney and reticuloendothelial system. Due to papain treatment, the Fab fragments lack the antigenic determinants of the Fc fragment resulting in reduced immunogenicity to patients as opposed to intact immunoglobulin products.71, 72, 75, 76, 78, 79, 84, 88, 89

Diseases treated

Digoxin immune Fab is indicated for patients with either life-threatening or potentially life-threatening digoxin toxicity or overdose.71, 79, 90, 91, 92, 93, 94, 95 Data from clinical trials have showed that both DigiFab and Digibind reduce levels of free digoxin in the serum to below the limit of assay quantitation for several hours after Fab administration.

Adverse reactions

Digoxin immune Fab (ovine) generally is well tolerated following intravenous (IV) administration.71, 72, 73, 76, 78 Hypokalemia may occur, sometimes developing rapidly in patients receiving digoxin immune Fab (ovine).71, 72, 79, 96, 97 DigiFab should not be administered to patients with a known history of hypersensitivity to papaya or papain unless the benefits outweigh the risks.

Immune Globulins: Antiinfectious

Hepatitis B immune globulin/HepaGam B/nabi-HB/BayHepB/HyperHEP B S/D

Description

Hepatitis B immune globulin (HBIG) is a specific immune globulin (hyperimmune globulin) that contains antibody to hepatitis B surface antigen (anti-HBs) prepared from plasma of healthy donors with high titer (>1:100,000) of anti-HBs antibody. It provides temporary passive immunity against hepatitis B virus (HBV).98, 99, 100, 101, 102, 103, 104

HepaGam-B is a solvent/detergent-treated sterile solution of purified gamma globulin containing antibody to HBs antigen that contains high titers of anti-HBs from plasma donated by healthy screened donors. Both HBIG and HepaGam-B are manufactured by a solvent/detergent (S/D) treatment procedure that is effective in inactivating lipid-enveloped viruses such as hepatitis B virus, hepatitis C virus, and human immunodeficiency virus type 1 and type 2. However, S/D is less effective against nonlipid-enveloped viruses such as hepatitis A virus and parvovirus B-19.100, 101, 104

Mechanisms of action

It provides passive immunization for individuals exposed to the hepatitis B virus by binding to the surface antigen and reducing rate of hepatitis B infection.

Diseases treated

HBIG provides passive prophylactic immunity to HBV infection for prevention of perinatal HBV infection in neonates born to HBs antigen-positive (HBsAg-positive) mothers,100, 101, 102, 103, 104, 105, 106 for postexposure prophylaxis in susceptible individuals exposed to HBV or HBsAg-positive materials (e.g., blood, plasma, serum),100, 101, 102, 103, 104, 107, 108, 109 sexual exposure to HBsAg-positive persons, for household exposure to persons with acute HBV infection, and for prevention of HBV recurrence in liver transplant recipients who are HBsAg-positive (HepaGam-B only).104, 110, 111, 112, 113, 114, 115, 116, 117 HBIG is not indicated for treatment of active hepatitis B infection and is ineffective in the treatment of chronic active hepatitis B infection.105

Adverse reactions

The local adverse reactions that may occur at the site of injection after intramuscular (IM) administration are pain, tenderness, swelling, and erythema.100, 101, 109 The systemic effects that may occur after IM administration are urticaria, angioedema, nausea, vomiting, myalgia, headache, flu- or cold-like symptoms, lightheadedness, and malaise have been reported.100, 101, 104

Varicella zoster immune globulin/VariZIG

Summary

Varicella zoster immune globulin (VZIG) is a specific immune globulin (hyperimmune globulin). VZIG is prepared from plasma of donors selected for high titers of antibodies to varicella zoster virus (anti-VZV) and used to provide temporary passive immunity against VZV.118, 119, 120

Mechanisms of action

VZIG acts by neutralizing varicella zoster virus via high titers of IgG antibodies present in the plasma used.

Diseases treated

VZIG is used for postexposure prophylaxis of varicella (chickenpox) in individuals who do not have evidence of varicella immunity and are at high risk for severe varicella infection and its complications. These high risk individuals include immunocompromised patients such as neonates whose mothers have signs and symptoms of varicella around the time of delivery (i.e., 5 days before to 2 days after), premature infants born at ≥28 weeks of gestation who are exposed during the neonatal period and whose mothers do not have evidence of immunity, premature infants born at <28 weeks of gestation or who weigh ≤1000 g at birth and were exposed during the neonatal period regardless of their mothers' evidence of immunity status, and finally pregnant women.118, 119, 121, 122

VZIG is now recommended for outbreak control and postexposure treatment, and the vaccine is available to children with humoral immunodeficiencies and selected children with HIV infection.122 Use of VZIG for postexposure prophylaxis in pregnant women exposed to VZV may prevent or reduce severity of varicella in the woman but does not prevent fetal infection.119, 121

VZIG is not indicated for individuals who previously received age-appropriate varicella vaccination and subsequently became immunocompromised because of disease or immunosuppressive therapy later in life. Bone marrow transplant recipients should be considered susceptible to varicella regardless of previous history of varicella or varicella vaccination in themselves or their donors. However, those who develop varicella or herpes zoster after transplantation should be considered immune to varicella.119

Adverse reactions

The most common adverse effects reported with VZIG in clinical trials in pregnant women, infants, and immunocompromised adults and children were injection site pain, headache, chills, fatigue, rash, and nausea. Severe hypersensitivity reactions may occur following administration of VZIG.118

Rimabotulinumtoxin B/Myobloc

Summary

Rimabotulinumtoxin B, a type B botulinum toxin produced by fermentation of the bacterium Clostridium botulinum type B (Bean strain), is a neuromuscular blocking agent (neurotoxin) and inhibitor of acetylcholine release at motor nerve terminals.123, 124, 125, 126

Mechanisms of action

Rimabotulinumtoxin B and other botulinum toxin serotypes act by inhibiting acetylcholine release at the neuromuscular junction via a three-step process, that is, toxin binding, toxin internalization, and inhibition of acetylcholine release into the neuromuscular junction leading to chemical denervation and flaccid paralysis.123, 124, 126, 127

Diseases treated

Rimabotulinumtoxin B is used for management of adults with cervical dystonia (also called as spasmodic torticollis) to reduce severity of abnormal head positioning and neck pain through reduction of undesired or excessive contraction of striated or smooth (involuntary) muscle.128, 129, 130, 131

Adverse reactions

The most common adverse effects reported with Botulinum toxin are dry mouth, dysphagia, dyspepsia, and injection site pain.123, 132, 133, 134 Serious hypersensitivity reactions have been rarely reported with onabotulinumtoxin A.127

Botulism immune globulin/BabyBIG

Summary

Botulism immune globulin IV (BIG-IV) is a specific immune globulin (hyperimmune globulin) that is prepared from plasma of adult volunteer donors immunized with pentavalent botulinum toxoid, which neutralizes free botulinum toxin types A and B. It is one of the most poisonous substances known and exists in seven antigenic variants (types A to G).120, 121, 135

Mechanisms of action

BIG-IV is a human-derived antitoxin that neutralizes botulinum toxin. BIG-IV has a half-life of approximately 28 days in vivo and large capacity to neutralize the toxin.135

Disease treated

Infant botulism occurs when young infants ingest spores of Clostridium botulinum that then germinate, colonize the GI tract, and produce botulinum toxin. This neurotoxin causes generalized weakness and loss of muscle tone. A single infusion will neutralize the toxin for at least 6 months and toxins type A or B that may be absorbed from the colon of an infant younger than 1 year old.121, 135, 136, 137, 138, 139

Adverse effect

Mild, transient, blush-like erythematous rash on the face or trunk occurred in 9%–14% of infants receiving BIG-IV in clinical studies.135, 140

Rabies immune globulin/bayrab/HyperRAB, imogam Rabies, KedRAB

Description

Rabies immune globulin (RIG) is a sterile solution of specific IgG that contains antibody to rabies antigen. It is used to provide temporary passive immunity to rabies infection as part of a postexposure prophylaxis regimen in unvaccinated individuals exposed to the disease or virus.141, 142, 143, 144

Mechanisms of action

RIG is a human-derived antitoxin that neutralizes rabies virus so that virus spread is reduced and its infective or pathogenic properties are inhibited. Specific rabies antibodies present in RIG neutralizes rabies. It should be used in conjunction with rabies vaccine and can be administered through the seventh day after the first dose of vaccine is given. RIG provides immediate, temporary rabies virus-neutralizing antibodies until the patient responds to active immunization and produces virus-neutralizing antibodies.121, 141, 142, 143, 144

Diseases treated

Given to all persons suspected of exposure to rabies with one exception, those who have been previously immunized with rabies vaccine and have a confirmed adequate rabies antibody titer should receive only vaccine.

Adverse reactions

Most common local adverse effects include tenderness, pain, muscle soreness, or stiffness that may occur at the site of injection. Low-grade fever, headache, and malaise may also occur.141, 142, 143

Immune Globulins: Immunomodulation

Rho(D) immune globulin/WinRho; RhoGam; Rhophylac, MicRhoGAM, BatRhoD, HyperRho

Summary

Rho(D) immune globulin (RhIG) consists of anti-Rho(D) IgG antibodies to the red blood cell Rho(D) antigen. RhIG is prepared from human pools of plasma of Rho(D)-negative donors immunized with Rho(D)-positive red blood cells after cold alcohol fractionation, and subsequent purification and infectious disease reduction technologies.145, 146, 147, 148, 149, 150

Mechanisms of action

The exact mechanism of action of Rho(D) immune globulin in the suppression of formation of anti-Rho(D) is not fully known.

In the treatment of preventing D alloimmunization, RhIG binds to Rho(D) antigen that entered the maternal circulation during fetal–maternal hemorrhage (FMH) involving an Rho(D)-positive fetus or transfusion with Rho(D)-positive blood, preventing stimulation of the mother's primary immune response to Rho(D) antigen. Therefore, by preventing the active production of anti-Rho (D) by the mother, the risk of hemolytic disease of the fetus and newborn in future pregnancies is decreased.145, 146, 147, 148, 149

In the treatment of idiopathic thrombocytopenic purpura (ITP), administration of Rho(D) immune globulin to Rho(D)-positive individuals is believed to cause transient mononuclear macrophage Fc receptor (FcR) blockade by complexes within the reticuloendothelial system, particularly the spleen, which spares the patient's IgG-coated platelets. This FcR blockade and decreased Fc-mediated phagocytosis of antibody-coated platelets result in increases of platelet counts in ITP patients.145, 149, 151, 152, 153, 154, 155, 156

Diseases treated

Prevent D alloimmunization in D-negative women of childbearing potential if the neonate is D+, weak-D positive, or D untested, and following perinatal events associated with FMH such as abortion, ectopic pregnancy, amniocentesis, chorionic villus sampling, external cephalic version, abdominal trauma, and antepartum hemorrhage. It is also used to prevent D alloimmunization in D-negative individuals who receive D+ blood components such as whole blood-derived platelets, apheresis platelets, and/or granulocytes. Similarly, it is used for the treatment of ITP in D+ patients who had not undergone splenectomy.145, 146, 147, 149, 151, 152, 153, 157, 158, 159, 160, 161, 162, 163, 164 Some preparations of Rho(D) immune globulin may be administered IM or IV (Rhophylac, WinRho SDF), whereas others are labeled for IM use only (MICRhoGAM, RhoGAM, HyperRHO S/D Full Dose, HyperRHO S/D Mini-Dose).145, 146, 147, 149, 165 When used for ITP treatment, RhIG must be administered IV.145, 149

Adverse reactions

Generally, mild with the most common being headache, fever, chills, pain at the injection site and, rarely, hypersensitivity reactions. Some degree of hemolysis is inevitable, but this is predictable and transient.146, 147

Immunoglobulin (generic)/bbrands: Bivigam, Carimune, Cuvitru, Flebogamma, Gammagard, GamaSTAN, Gammaked, Gammaplex, Gamunex-C, Hizentra, Hyqvia, Octagam Privigen

Summary

Immune globulin IM (IMIG), immune globulin IV (IVIG), and immune globulin subcutaneous are sterile, nonpyrogenic preparations of globulins containing many antibodies normally present in adult human blood. Immune globulins (IG) are collected either by whole blood donations as recovered plasma (20%), or by apheresis as source plasma (80%). IVIG is a highly purified product consisting mostly of IgG with a half-life of 21–28 days.

Hyperimmune globulin (Hyper-Ig) products are manufactured from donors with high Ig titers with specificity to antigenic determinant(s) of interest. High titers of these donors can be achieved by natural immunity, prophylactic immunizations, or through targeted immunizations. Hyper-Ig products should contain at least fivefold-increased titers compared to standard preparations of IVIG.

IVIG production is regulated by the IUIS/WHO (International Union of Immunological Societies/World Health Organization), which require the following:

  • Source material must be plasma obtained from a minimum pool of 10,000 donors;

  • Product must be free of prekallikrein activator, kinins, plasmin, preservatives, or other potentially harmful contaminants;

  • IgA content and IgG aggregate levels need to be as low as possible;

  • Product must contain at least 90% intact IgG;

  • IgG should maintain opsonin activity, complement binding, and other biological activities;

  • IgG subclasses should be present in similar proportions to those in normal pooled plasma;

  • Antibody levels against at least two species of bacteria (or toxins) and two viruses should be determined;

  • Product must demonstrate at least 0.1 international units of hepatitis B antibody per mL, and hepatitis A radioimmunoassay titer of at least 1:1000;

  • Manufacturer should specify the contents of the final product, including the diluent and other additives, and any chemical modification of IgG.166, 167, 168, 169, 170, 171, 172

Mechanisms of action

The mechanisms of Ig-induced immunomodulation are incompletely understood but include macrophage Fc receptor blockage by immune complexes formed between IVIG and native antibodies, modulation of complement, suppression of antibody production, suppression of inflammatory cytokines and chemokines, and/or antiidiotypic regulation of autoreactive B-lymphocytes or antibodies.

As IVIG contains a diverse group of antibody specificities, which protects recipients against multiple infections by eliminating opsonized infectious organisms via antibody-dependent cell-mediated cytotoxicity or by complement activation. This is followed by lysis and/or neutralization of soluble infectious proteins by immune complex formation and elimination through the RES.166, 170, 173, 174, 175

Diseases treated

IVIG is indicated for the treatment of primary immune deficiency, secondary immune deficiency, ITP, Kawasaki disease, and congenital hypogammaglobulinemia. Currently, there is an extensive list of diseases for which IVIG could be used. It also has immunomodulatory properties resulting in an increasing list of both FDA-approved and nonapproved indications.

IMIG is used to provide passive immunity to hepatitis A virus infection for preexposure or postexposure prophylaxis in susceptible individuals who are at risk of or have been exposed to the virus. IMIG and IVIG are used to prevent or modify symptoms of measles (rubeola) in susceptible individuals exposed to the disease <6 days. IVIG is used for replacement therapy to promote passive immunity in patients with primary humoral immunodeficiency who are unable to produce sufficient amounts of IgG antibodies and in the management of ITP to increase platelet counts, to prevent and/or control bleeding, or to allow these patients to undergo surgery.

IVIG is used for prevention of bacterial infections in patients with hypogammaglobulinemia and/or recurrent bacterial infections associated with B-cell Chronic Lymphocytic Leukemia. IVIG is used in conjunction with aspirin therapy for initial treatment of the acute phase of Kawasaki disease. IVIG is also used to treat chronic inflammatory demyelinating polyneuropathy to improve neuromuscular disability and impairment, and for maintenance therapy to prevent relapse. Furthermore, IVIG is used for maintenance treatment to improve muscle strength and disability in adults with multifocal motor neuropathy.166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201

Adverse reactions

Approximately 2%–10% of infusions are associated with adverse reactions that include those at the infusion site (erythema, pain, swelling, pruritus, heat), phlebitis, eczema, fever, chills, myalgias, malaise, flushing, rash, diaphoresis, pruritus, bronchospasm, chest pain, back pain, extremity pain, dizziness, blood pressure changes, nausea, vomiting, and headache.167, 170, 172, 177, 178, 179, 181, 182, 183

Passive Monoclonal Antibody Treatment

In the late 20th century (∼1986), monoclonal antibodies were developed. The first monoclonal antibodies (Mabs) were of xenographic source and were wrought with problems of immunogenicity. These early Mabs did not gain favor until chimerization took pace in the mid-1990s, and in 1998 two Mabs were approved to treat one respiratory syncytial virus and the other certain breast cancers. Further development to humanize and then generate fully human Mab led to an evolution of therapies utilizing these agents. Mabs are being researched or approved to treat a multitude of diseases that include oncologic, inflammatory, autoimmune, cardiovascular, respiratory, neurologic, allergic, benign hematologic, infectious, orthopedic, coagulopathic, and metabolic indications and to decrease disease morbidity (diminution of pain), modify disease progression (i.e., macular degeneration, diabetes), and potentially alter anatomic development. In this section of the chapter, we will review the history of use of these passive monospecific antibody therapies, their mechanism of action, pharmacologic-therapeutic classification, particular medical indication, adverse reactions, and potential future use of these medications.201

Mechanism of action

Depending on the antigenic target of these antibodies multiple events are set into action. Immunologic changes occur as the specific antigens are presented more efficiently to effector cells. Some of these actions create decreased inflammatory and allergic responses, while other effects generate antibody-dependent cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Other actions can block receptor interaction with ligands by either binding with ligands or their cognate receptors (i.e., allow activation of NK cells). Interactions may also directly cause initiation of programmed cell death (apoptosis), cessation of growth/replication/proliferation, or lead to changes in metabolism. Moreover, there are also antibodies against infectious agents to prevent cell adhesion for entry, spread, replication, and contagion. Antibodies may also be directed against toxins leading to various methods of inactivation.201

Adverse reactions

Depending on their mode of action, Mabs are associated with a myriad of side effects. They can be associated with immunogenicity that can cause a decrease in their effectiveness. Antineoplastic antibodies can be associated with tumor lysis syndrome. Similarly, reactivation of underlying infections can occur leading to progressive multifocal leukoencephalopathy, HBV, fungal, parasitic, or tuberculosis infections. Other adverse reactions include but are not limited to initiation of autoimmune disorders, increased risk for malignancy, cardiac arrhythmia, angina/ischemia, cytopenias, hemorrhage, and allergic reactions including anaphylaxis, embryo–fetal toxicity (if can cross placental barrier), and even death.

Types of Illness Treated

Oncology

Malignancies can be caused by infectious agents, toxins, or genetic mutations with changes in control of growth, proliferation, or programmed cell death. Historically these have been treated with a variety of radiation therapies to eradicate malignant cells or with chemotherapeutic agents to enhance maturity, decrease proliferation, or cause destruction of cancer cells. In some cases intense high-dose chemotherapy is used to cause cancer remission with stem cell transplants for subsequent rescue. Passive antibody therapy may replace or be additive to other pharmacology therapies and increase chances for complete remission, prolong disease-free survival, and overall survival.

B-cell chronic lymphocytic leukemia

Rituximab (Rituxan) is a chimeric murine/human Mab (IgG1κ) that binds to CD20 (human B-lymphocyte-restricted differentiation antigen, Bp35 {controlling differentiation and possible calcium ion channel}). Its mechanism of action is not entirely clear and may involve CDC and ADCC. Many studies have shown this antibody to have an additive benefit to standard chemotherapy alone. This antibody has been approved by the FDA to treat chronic lymphocytic leukemia (CLL) since 1997. Nowadays, this medication is often combined with ibritumomab in treating CLL (to be discussed with non-Hodgkin's lymphoma).202, 203

Alemtuzumab (Campath) binds to CD52 and is a humanized rat Mab (IgG1κ) binding to receptors on both T and B cells as well as macrophages, NK cells, and neutrophils, leading to CDC and ADCC. The resultant cytopenias lead to a severe immunocompromised state. Alemtuzumab was FDA approved as a single agent in the treatment of B-cell CLL in 2001.204

Ofatumumab (Ocrevus) is a human Mab (IgG1κ) with CDC that binds to CD20 near the cellular membrane. In phase II studies, this agent had 86% objective response rate (ORR) when used alone and with CHOP therapy had 100% ORR and 62% complete remission (CR); whereas, in phase III trials, this Mab showed ORR of 10% after rituximab relapse. This medication was approved by the FDA to treat CLL in 2009.205

Monalizumab is a humanized Mab (IgG4κ) that binds to CD94/NKG2A (an inhibitory signal receptor transmitter) on NK cells. Monalizumab demonstrated blockade of NKG2A/HLA-E and restores the ability of NK cells to lyse B cells in vitro. In addition, this Mab was shown to be of benefit in murine models. Ongoing phase I/II studies will be completed in 2019.206

Otlertuzumab is a humanized Mab fragment (IgG Fab’) with specificity to CD37 that induces both ADCC and caspase-independent apoptosis. In a phase II study both better progression-free survival (PFS) and ORR were observed when used with bendamustine compared to bendamustine used alone.207

Urelumab is a human Mab (IgG4κ) with specificity to CD134 (an immune checkpoint inhibitor). This antibody has completed safety phase I dosing trials. Higher doses lead to significant hepatotoxicity. Safe dosing is now established in clinical phase II studies to be completed in 2020.208, 209

Ulocuplumab is a human Mab with specificity to CD184 (CXCR4). In vitro studies showed apoptotic effects via production of oxygen species that was not associated with better caspase activation than AMD3100. Phase I studies were completed in 2014, no manuscripts were found for review. This medication is presently in phase II trials against acute myelocytic leukemia (AML) to be completed in 2021.210, 211

Other monoclonal antibodies not demonstrating benefit in clinical trials for CLL include apolizumab, dacetuzumab, and gomiliximab (aka lumiliximab)212, 213, 214, 215

Acute myelocytic leukemia

AML is the leading cause of leukemic mortality in the United States (US). Over the last 10 years therapy has not changed significantly for this disease. Novel therapies have been developed in the last decade, some showing temporal success and some showing a brighter tomorrow.216

AMG330 is a bispecific T-cell engager (BiTE) antibody with specificity for CD3 and CD33. This Mab is currently in clinical trials to be completed in 2020 for treatment of AML. A BiTE antibody stimulates ADCC (via T cells) in the presence of antigenic targets on cells of interest. In vitro studies have shown effective lysis of AML cells, while in animal studies it has demonstrated significant decrease in tumor burden.217

IMGN632 is an anti-CD123 antibody complexed to a DNA mono-alkylating agent. In vitro studies showed it had more potency against AML cells than to normal myeloid progenitor cells. In animal models there was an excellent response rate against tumor cells. Ongoing clinical trials will be completed in 2021.216

Talacotuzumab is a humanized monoclonal antibody (IgG1-2κ) with specificity to interleukin (IL)-3 receptor subunit-α (CD123, a growth and differentiating receptor). This antibody induces ADCC both in vitro and in animal models. Phase III clinical trials were reportedly completed in 2018; published results are forthcoming.218

Samalizumab is a humanized Mab (IgG2/IgG4κ) with specificity to CD200 (OX-2membrane glycoprotein) is in phase II trials to be completed in 2021.219

Ficlatuzumab is a humanized Mab (IgG1κ) in a phase I trial to treat refractory/relapsing AML to be completed in 2020.220

Other Mab not demonstrating benefit in clinical trials or withdrawn following postmarketing for AML include gemtuzumab ozogamicin (FDA approved 2000 withdrawn 2010 secondary to venoocclusive disease) and lintuzumab (no added benefit over standard chemotherapy).221, 222, 223

Multiple Myeloma

Daratumumab (Darzalex) is a human Mab (IgG1κ) with specificity to CD38 (functions reportedly include receptor-mediated adhesion and signaling events, as well as important bifunctional ectoenzymatic activities that contribute to intracellular calcium mobilization. This Mab mechanism of action is thought to induce CDC, ADCC, antibody-dependent cellular phagocytosis, and apoptosis. This medication is used to treat refractory and recurrent multiple myeloma.224, 225

Silutuximab (Sylvant) is a chimeric Mab (IgG1κ) with specificity to IL-6. This medication was FDA approved in 2014 for multicentric Castleman's disease (MCD) with HIV negative and HHV-8 negative. There are ongoing studies in phase II clinical trials to be completed in 2019.226, 227

B-cell acute lymphoblastic leukemia (B-cell ALL)

Blinatumomab (Blincyto) is a mouse double heavy-chain fragment (Murine {scFv - kappa – heavy} – {scFv - heavy – kappa}) with specificity for CD19 and CD3 known as a BiTE. This Mab's mode of action is by directing CD3+ effector memory T cells to CD19+ target cells leading to T-cell activation and B-cell apoptosis. This biologic is used to treat relapsed/refractory cell ALL. In phase III trials event-free survival almost tripled and duration of remission almost doubled.228, 229, 230

Hodgkin's lymphoma

Hodgkin's lymphoma is a rare malignancy affecting young adults with a peak incidence in patients >55 years old. Up to 40% of these patients can develop relapsing disease. Brentuximab vedotin (Adcentrix) is a chimeric humanized Mab drug conjugate (Mab + linker + payload {IgG1κ +protease cleavage linker + monomethyl auristatin E [MMAE]}) with specificity to CD30 (a cell membrane protein of the tumor necrosis factor receptor superfamily member 8. MMAE is a microtubule-disrupting agent. The combination of this a Mab and drug conjugate disrupts the intracellular microtubule network causing cell cycle arrest at G2/M stage and apoptosis. This medication has a 43% PFS at 30 months.231

Mab to look out for in the future include Camidanlumab tesirine (ADCT-301) a human Mab (IgG1κ). This Mab has specificity to CD25 (a IL-2 receptor alpha subunit) with a drug conjugate. The drug is released intracellularly and causes DNA interstrand crosslinks. This Mab is in phase I studies to be completed in 2019 for Hodgkin's and non-Hodgkin's T- and B-cell lymphomas. In addition, there are clinical phase I studies against multiple solid tumors to be completed in 2021.232, 233

Agents abandoned or not found to be beneficial include apolizumab, denintuzumab mafodotin (HBU-12), iratumumab (MDX060), and lucatumumab (HCD122).212, 234, 235

Anaplastic large cell lymphoma

Brentuximab vedotin (Adcentrix) is an FDA-approved medication for patients with refractory or relapsed anaplastic large cell lymphoma who achieved CR. This Mab had 79% OS and 57% PFS at 5 years, with median response duration not reached at time of publication.236

Breast Cancer

Atezolizumab (Tecentriq) is an FcγR binding–deficient, fully humanized Mab (IgG1κ). This Mab binds to programmed death ligand I (PD-L1) to prevent interaction with receptors PD-1 and B7.1 (a costimulatory cell-surface protein), reversing T-cell suppression. Activation of B7.1 can potentially stimulate long-term responses through development of new immunity via priming and activation of T cells in lymph nodes. A lack of FcγR binding decreases ADCC of the T cells enabling more tumor-specific T cell to remain active. This medication was approved by the FDA in 2019 to treat triple negative (estrogen receptor, progesterone receptor, human epidermal growth factor receptor-2) unresectable or metastatic breast cancers.237, 238

Colorectal Cancer

Bevacizumab (Avastin) is a humanized Mab (IgG1κ) with specificity to vascular endothelial growth factor-a (VEGF-A) that acts as an inhibitor of angiogenesis. It was FDA approved for treatment of colorectal cancer and has recently been approved for multiple other cancers including ovarian, fallopian cancers, renal cell carcinoma, and recurrent glioblastoma multiforme (GBM).239, 240

Urothelial Carcinoma

Atezolizumab (Tecentriq) is FDA approved as a single agent in urothelial carcinoma and for patients with disease progression despite other chemotherapy treatment.241, 242

Nonsmall cell lung cancer

Atezolizumab (Tecentriq) is FDA approved as a single agent for nonsmall cell lung cancer (NSCLC).

Bevacizumab (Avastin) is FDA approved for treatment of locally advanced, recurrent or metastatic, nonsquamous NSCLC.

Nivolumab (Opdivo) is an FDA-approved human Mab (IgG4κ) immunoglobulin and blocks PD-1 preventing interaction PD-1 and its ligands PD-L1 and PD-L2. It is used to treat RCC, NSCLC, Hodgkin's lymphoma, melanoma, small cell lung cancer, colorectal cancer, and squamous cell carcinoma of the head and neck. In phase III clinical trials, nivolumab performed better than docetaxel in the treatment of NSCLC.243, 244, 245

Ovarian/cervical fallopian cancer

Bevacizumab (Avastin) is FDA approved for treatment of locally advanced, recurrent or metastatic, ovarian, cervical, and fallopian cancers after treatment with chemotherapy regimens and surgery.246

Merkel Cell Carcinoma

Merkel cell carcinoma is a rare aggressive cutaneous malignancy caused by infection with polyoma virus and exposure to ultraviolet radiation. This cancer was classically treated with chemotherapeutic agents leading to rare durable responses. Avelumab (Bavencio) is a fully human Mab (IgG1λ) with specificity to PD-L1. This Mab was approved by the FDA for treatment of Merkel cell carcinoma in 2017. Treatment with this Mab increases response rates to about 50% and extended durable response times approximately five times.247, 248 This Mab is in clinical trial to treat other solid tumors including but not limited to hepatocellular, ovarian, esophagogastric, colorectal NSCLC, testicular, urothelial, and adrenocortical carcinomas.249

Neuroblastoma

Neuroblastoma is an aggressive tumor of children with a 5-year survival of about 50%. Treatment classically is high-dose intensive chemotherapy, myeloablative chemotherapy with stem cell rescue, and/or irradiation therapy. Dinutuximab (Unituxin) is a chimeric Mab (IgG1κ) with specificity to GD2 ganglioside that has mechanisms of action via CDC and ADCC. This Mab is used in patients who have had at least a partial response to classic therapy.250, 251

Glioblastoma Multiforme

GBM is the most common malignant primary brain tumor in adults. This disease remains incurable.

Bevacizumab (Avastin) is FDA approved for treatment of recurrent GBM as salvage therapy. This medication with chemotherapy increases overall survival by 4 months but as a single agent is not effective.252

Relatlimab (BMS-986,016) is a human Mab (IgG4κ) with specificity to lymphocyte activation gene 3 (LAG3, CD223) and is in phase I clinical trials to be completed in 2020 for treatment of GBM.253

Tanibirumab (aka Olinvacimab, TTAC-0001) is a human Mab (IgG1) with specificity to vascular endothelial growth factor receptor-2 (VEFR-2) and is in phase II studies to treat GBM to be completed in 2020.254, 255, 256

Malignant Ascites

Catumaxomab (Removab) is a trifunctional rat/murine hybrid antibody (IgG2a/IgG2b). Catumaxomab consists of one “half” (one heavy chain and one light chain) of an antiepithelial cell adhesion molecule (anti-EpCAM) antibody and one-half of an anti-CD3 antibody, so that each molecule of catumaxomab can bind both EpCAM and CD3. In addition, the Fc-region can bind to an Fc receptor on accessory cells such as other antibodies, which has led to calling the drug a trifunctional antibody. This antibody's mechanism of action is through ADCC. It is approved for use in Europe for malignant ascites from ovarian, gastric, colon, pancreatic, breast, and endometrial carcinoma and is a pending review for approval by the FDA.257, 258, 259, 260

Cutaneous squamous cell carcinoma

Cemiplimab (Libtayo) is a human Mab (IgG4) for treatment of cutaneous squamous cell carcinoma (CSCC) that is metastatic or locally advanced and not amenable to surgery. CSCC is second only to basal cell carcinoma as the most common skin cancer. Surgical intervention is not possible in 5% of patients. This Mab offers a treatment with less morbidity than palliative radiation or surgery, and gives an ORR in 50% of these otherwise untreatable patients. There are many additional phase II studies involving this Mab to be completed from 2020 to 23.261, 262

Autoimmune/Inflammatory Diseases

Inflammatory Bowel Disease

Inflammatory bowel disease (IBD) pathophysiology remains unknown but may have genetic, infectious, autoimmune origins including cell-mediated immunity. These diseases may be classified as ulcerative colitis (UC), isolated to the colon, or Crohn's disease primarily found in the colon but may involve the entire gastrointestinal tract. With long-standing active disease, malignancy is much more frequent in UC than in Crohn's disease. Mild UC is treated with antiinflammatory agents such as sulfasalazine and glucocorticosteroids. For more severe disease, high-dose steroids may be used to maintain disease quiescent and low-dose steroids to keep disease in remission. Low-dose chemotherapeutic agent or immunosuppressive agent may also be added if dose of corticosteroids is too high to maintain remission. Surgery may be necessary to control disease. For Crohn's disease, medical therapy is usually less successful in managing the disease and surgery may be necessary but is not curative as in UC. For both of these disease processes, passive antibody therapy may offer not only control of disease but possible complete remission from mucosal damage.263, 264

Adalimumab (two formulations: Humira and Amjevita) is a recombinant human Mab (IgG1) with specificity to tumor necrosis factor alpha (TNF-α). Both forms are FDA approved to treat Crohn's disease as well as multiple types of rheumatoid arthritis. In Crohn's disease, this medication decreases signs and symptoms of disease and is able to induce clinical remissions.265, 266

Certolizumab (Cimzia) is a recombinant humanized m fragment with TNF-α as target. It is FDA approved for both Crohn's disease and Rheumatoid arthritis.267, 268, 269

Vedolizumab (Entyvio) is a humanized Mab (IgG1κ) that has selectivity for integrin α4β7 and is FDA approved for treatment of Crohn's disease. This Mab mode of action is to selectively block trafficking of memory T cells into inflamed gut tissue by inhibiting α4β7-mucosal addressin cell adhesion molecule-1 (MAd-CAM-1) interaction with intestinal vasculature. This medication has shown a good safety profile with no cases of promyelocytic leukemia (PML), no increased risk of infections, malignancies compared with classically treated IBD, and low incidence of infusion-related reactions. This medication is also FDA approved for UC.270, 271

Infliximab (Remicade, Inflectra, Remsira) is a chimeric Mab (IgG1κ) with specificity to TNF-α and is FDA approved for IBD and multiple inflammatory arthritic diseases. This medication allows for steroid-free remission within months of starting therapy.272

Natalizumab (Tysabri) is a humanized Mab (IgG2κ) with selectivity to CD62L (L selectin α4 subunit of α4β1 and α4β7 integrins of leukocytes, not neutrophils, VLA-4). This Mab is FDA approved for Crohn's disease and multiple sclerosis. This medications is effective in induction of clinical remission in moderate-to-severe Crohn's disease. This medication does have the risk of PML.273, 274

Other Mab being studied for Crohn's disease but not yet approved by the FDA include Ustekinumab, brazikumab, etrolizumab, risankizumab, and ontamalimab. In contrast, Mabs studied but not beneficial for Crohn's disease include andecaliximab, eldelumab, and fontolizumab. Refer to Table 16.1 .

Table 16.1.

Summary of Monoclonal Antibody Therapies.

Generic Drug Name Brand Name Type of Antibody AHFS Classification Dosage Form(s) Target
8H9 Iodine 124 monoclonal antibody
(Murine)
Antineoplastic
Neuroblastoma, sarcoma, metastatic brain cancers
Another study Sloan Kettering using I331 version phase I good results
Intravenous B7–H3
Abagovomab Monoclonal antibody
(Murine)
An antiidiotypic mAb that mimics ovarian cancer CA125 protein
Antineoplastic
Phase II study for ovarian cancer
Phase III good immune response but no increase RFS or OS no benefit
Subcutaneous CA-125
Abatacept Orencia
FDA 2005
EU 2010
Recombinant soluble fusion protein of the extracellular domain of human cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) linked to the modified Fc portion of human immunoglobulin G1 (IgG1). Disease modifying
Rheumatoid arthritis
Juvenile and adult psoriatic arthritis (phase III)
Subcutaneous or intravenous Selective costimulation modulator, inhibits T cell (T lymphocyte) activation by binding to CD80 and CD86, thereby blocking interaction with CD28. This interaction provides a costimulatory signal necessary for full activation of T lymphocytes.
Abciximab
c7Ec Fab
ReoPro
FDA 1994
EU 1995 (country-specific approval)
Human-murine chimera
Recombinant monoclonal IgG1 Fab
Procedure modification
High-risk coronary intervention
Platelet aggregation inhibitor
Intravenous Platelet glycoprotein IIb/IIIa receptor (CD41 7E3)/Intergrin α-IIb
Abituzumab
DI17E6
EMD525797
Humanized monoclonal antibody IgG2κ Antineoplastic
Colorectal cancer phase I 2013, phase II 2015 primary endpoint PFS not met
Sclerosing interstitial lung disease phase II terminated 2018 slow enrollment
Prostate phase IIno significant increase PFS
Intravenous CD51 (?integrin alpha V)
Abrilumab
AMG 181
Phase II study discontinued development (2016) Integrin α-4 β-7
Actoxumab Human monoclonal antibody Disease modifying
Clostridium difficile
Phase I and II anti-CDTB1 much better
Clostridium difficile toxin A
Adalimumab Humira
FDA 2002
EU 2003
Amjevita
FDA 2016
EU 2017
Recombinant human IgG1 monoclonal antibody Disease modifying
Humira
Rheumatoidarthritis; juvenile idiopathic arthritis; psoriatic arthritis; ankylosing spondylitis; Crohn's disease, plaque psoriasis
Amjevita
Arthritis; juvenile rheumatoid arthritis; psoriatic arthritis; rheumatoid colitis; ulcerative Crohn's disease; psoriasis; spondylitis; ankylosing
Possibly hemolytic disease of newborn
Injection subcutaneous TNF-α
Adecatumumab
MT-201
Recombinant human monoclonal antibody IgG1κ Antineoplastic
Breast phase Ib+, colorectal and prostate
Phase II completed
Phase III soon?
Intravenous EpCAM (CD326) epithelial cell adhesion molecule
Aducanumab Human monoclonal antibody IgG1 Disease modifying
Alzheimer's disease
Phase III x 2 ongoing started 2015
Intravenous Beta-amyloid (N-terminus 3–6) soluble oligomers and insoluble fibrils
Afasevikumab Human monoclonal antibody IgG1κ Disease modifying
Multiple sclerosis
Phase I completed
Nothing in pubmed
Subcutaneous IL17A and IL17F
Afelimomab Murine F(ab')
Antibody Fab’ fragment IgG3κ
Disease modifying
Sepsis
Phase III trial marginal benefit abandoned
TNF-α
Alacizumab pegol Humanized monoclonal antibody F(ab')2 Limited information on development;
Cancer
VEGFR2
Alemtuzumab
LDP-03
Campath-1H
Lemtrada
FDA 2014
EU 2013
MS
Campath
FDA 2001
EU 2001
CLL
Humanized rat monoclonal antibody IgG1κ Antineoplastic
B-Cell CLL, CTCL, T cell lymphoma
Disease modifying
Multiple sclerosis (phase III)
Not effective for kidney transplant conditioning or rejection prevention
Intravenous CD52
Alirocumab Praluent
FDA 2015
EU 2015
Human monoclonal antibody IgG1 Disease modifying
Decrease cholesterol
Phase III
Subcutaneous Proprotein convertase subtilisin kexin type 9 (PCSK9)
Altumomab pentetate In98 Hybri-ceaker Murine monoclonal antibody IgG1 Diagnostic purpose radiology colorectal cancer (diagnosis) CEA
ALX-0171 Trimeric nanobody Antiinfectious
RSV phase II 2020
Inhalation RSVF
Amatuximab
MORAb-009
Chimeric murine-human monoclonal antibody IgG1κ Antineoplastic
Ovarian cancer
Phase II
Now research on using to treat mesotheliomas
Phase I/II
Pancreatic cancer
Intravenous Mesothelin
Prohibits binding of MSLN with antigen CA125/MUC16
AMG330 Bispecific T-cell engager (BiTE) Antineoplastic
AML phase I AML 2020
Intravenous CD33 and CD3
Anatumomab mafenatox Murine monoclonal fragment Fab Antineoplastic
Nonsmall cell lung carcinoma
Tumor-associated glycoprotein 72 (TAG-72)
Andecaliximab
GS 5745
Chimeric monoclonal antibody IgG4κ Antineoplastic gastric cancer phase I, II, III ongoing or gastroesophageal junction adenocarcinoma phase III ongoing
Crohn phase II no response, UC
Intravenous Gelatinase B is a matrix metalloproteinase-9 (MMP-9)
Anetumab ravtansine
In98
Human monoclonal antibody IgG1λ Antineoplastic ovarian phase II, lung, pancreatic phase I, breast now research on using to treat mesotheliomas
Phase II
Cervical cancer ?preclinical
Mesothelin
Prohibits binding of MSLN with antigen CA125/MUC16
Anifrolumab Human monoclonal antibody IgG1κ Disease modifying
Systemic lupus erythematosus phase I and IIb 2018
Intravenous Interferon α/β receptor
Anrukinzumab (=IMA-638) Humanized monoclonal antibody IgG1κ Disease modifying
Asthma phase II ?results
UC phase II no benefit
IL-13
Apolizumab Humanized monoclonal antibody Antineoplastic non-Hodgkin's lymphoma abandoned 2009 toxic effects
2009 CLL phase I/II
HLA-DRβ
Arcitumomab CEA-Scan
FDA 1996
EU 1996
Withdrawn EU market 2005
Murine monoclonal antibody IgG1 Fab' Diagnostic imaging
Gastrointestinal cancers
Colorectal cancers
CEA
Ascrinvacumab Human monoclonal antibody Antineoplastic mesothelioma
Nothing in pub med or web search
Activin receptor-like kinase 1
Aselizumab Humanized monoclonal antibody Disease modifying
Severe injured patients phase II 2004, no benefit
L-selectin (CD62L)
Atezolizumab
MPDL3280A
Tecentriq
FDA 2016
Fc engineered, humanized monoclonal antibody IgG1κ Antineoplastic agent, treat metastatic urothelial carcinoma, non-small cell lung cancer
Phase III
Bladder/urothelial cancer phase I
Breast cancer phase Ib triple marker neg breast cancer
Intravenous Binds to PD-L1 and blocks interactions with the PD-1 and B7.1 receptors FDA-approved atezolizumab (TECENTRIQ, Genentech, Inc.), in combination with bevacizumab, paclitaxel, and carboplatin for the first-line treatment of patients with metastatic nonsquamous, nonsmall cell lung cancer (NSq NSCLC) with no EGFR or ALK genomic tumor aberrations
Atidortoxumab Human monoclonal antibody IgG1κ Limited information on use and development Negative search PubMed-internet Staph aureus alpha toxin
Atinumab Human monoclonal antibody IgG4κ Disease modifying
Acute spinal cord injury
RTN4
Atorolimumab Developed?? Human monoclonal antibody IgG3 Disease modifying hemolytic disease of the newborn Rhesus factor
Avelumab Bavencio
FDA 2017
Human monoclonal antibody IgG1λ Antineoplastic
Cancers, ovarian, gastric, nonsmall cell lung (NSCLC), metastatic, solid tumors
phase II
Studies completed metastatic Merkel cell carcinoma
Intravenous PD-L1
Azintuxizumab vedotin Chimeric/humanized monoclonal antibody IgG1 Antineoplastic
Nothing in PubMed
CD319
BAN-2401 Humanized monoclonal antibody IgG1 Disease modifying Alzheimer A phase IIb study ongoing started 2013 Intravenous Soluble Aβ amyloid protofibrils
Bapineuzumab Humanized IgG1 monoclonal antibody Disease modifying
Alzheimer's disease
Phase III no more studies discontinued research 2012 ARIA-E, amyloid-related imaging abnormalities–edema
Intravenous Beta amyloid
Fibrillary and soluble β amyloid
Basiliximab Simulect
FDA 1998
EU 1998
Chimeric monoclonal antibody IgG1κ Immunosuppressive agents
Prophylaxisof acute rejection in allogeneic renal transplantation
Intravenous CD25 (α chain of IL-2 receptor)
Bavituximab Chimeric monoclonal antibody IgG1κ
IgG3 (SUNRISE trial)
Cancer, viral infections (Hep C) phase III NSCLC failed to improve survival Sunrise trial stopped Feb 2016, phase II/III breast cancer, phase II pancreatic cancer, phase I/II trial hepatocellular carcinoma, phase I malignant melanoma + rectal cancer good response rectal; not for prostate cancer, phase II hepatitis C not resulted? Phosphatidylserine
BAY-103356
CDP 571
Humicade
Senlizumab
Humanized monoclonal antibody IgG4κ For research only
Phase II 1995
TNF α
BCD-100 Human monoclonal antibody Antineoplastic
Phase II/III melanoma
NCT03269565 (complete Dec 2019)
Intravenous Programmed cell death-1 (PD1)
Bectumomab LymphoScan Fab’-IgG2κ Antineoplastic
Non-Hodgkin's lymphoma (detection)
CD22
Begelomab Begedina Murine IgG2b Disease modifying GvHD phase II/III DPP4 binds CD26 on T lymphocytes
Belantamab mafodotin Humanized monoclonal antibodymab Antineoplastic No studies or info on clinical trial, PubMed, FDA substance BCMA
Belatacept Nulojix
FDA 2011
Soluble fusion
Protein consisting of the modified extracellular domain of CTLA-4 fused to Fc domain of a recombinant human monoclonal antibody IgG1
Immunosuppressive agents
Prophylaxisrenal transplant rejection in adults
Phase III
FDA approved
Intravenous Selectively inhibits T-cell activation through costimulation blockade binds to both CD80 and CD86 blocking CD28
Belimumab Benlysta
FDA 2011
EU 2011 LymphoStat-B
Human monoclonal antibody IgG1λ Disease modifying
Kidney transplant phase II
Treat SLE (testing phase III for renal involvement)
Phase II Rheum arthritis failure
Phase II Srogren ±
GVHD ongoing
Intravenous
Subcutaneous
B-cell activating factor (BAFF), B-lymphocyte stimulator
Bemarituzumab Humanized monoclonal antibody Antineoplastic FGFR2
Benralizumab Fasenra
FDA 2017
EU 2017
Humanized monoclonal antibody IgG1κ Disease-Modifying
Asthma phase III completed
Severe asthma eosinophilic subtype
Subcutaneous Interleukin-5 (IL-5α) receptor alpha subunit-directed cytolytic (CD125)
Berlimatoxumab Human monoclonal antibody Staph aureus bicomponent leukocidin No studies clinical, no find creative, zero pub med
Bermekimab
MABp1
T2-18C3
CA-18C3
Xilonix Human monoclonal antibody IgG1κ Disease modifying psoriasis phase III x2 2020
Ank spond II 2022
Psor arth II 2020 III 2020
Subcutaneous
Intravenous
IL17A
Bersanlimab Human monoclonal antibody ICAM-1
Bertilimumab CAT-214 Human monoclonal antibody IgG4κ Disease modifying
Severe allergic disorders phase II atopic dermititis
Ongoing studies bullous pemphigoid and ulcerative colitis phase II
Intravenous CCL11 (eotaxin-1)
Besilesomab Scintimun
EU 2010
Not FDA approved
Murine monoclonal antibody IgG1κ Diagnostic use
Inflammatory lesions and metastases (detection)
CEA-CAM8-related antigen
Bevacizumab Avastin
FDA 2004
EU 2005
Humanized monoclonal antibody IgG1κ
BiTE
Antineoplastic agent
Antiangiogenesis inhibitor
Colorectalcancer 2004, NSCLC 2006, RCC 2009, GBM phase III, ovarian cancer, metastatic cervical cancer, fallopian 2014
Breast cancer (FDA removed approval for breast cancer 2010)
Recurrentglioblastoma multiform
Nonsquamous nonsmall cell lung cancer
Intravenous solution or ophthalmic injection
May not be so good for GBM or ovarian
VEGF-A anti-angiogenesis inhibitor
Bezlotoxumab Zinplava
FDA 2016
EU 2017
Human monoclonal antibody IgG1 Disease modifying phase III studies done MODIFY I and II
Modify III ongoing
Pseudomembranous colitis
Intravenous Clostridium difficile colitis anti-B toxin
Biciromab FibriScint Murine monoclonal fragment Fab’ IgG1κ Detect cardiovascular thromboembolism (diagnosis) Fibrin II, beta chain
Bimagrumab
BYM338
Human monoclonal antibody IgG1λ Disease modifying
Myostatin inhibitor
DM II decrease BMI phase II
Sporadic inclusion body myositis phase III not meet endpoint
Treat sarcopenia in older adults phase II
Intravenous Activin A receptor type IIB (ACVR2B)
Bimekizumab Humanized monoclonal antibody IgG1κ Disease modifying
Ankylosing spondylitis (2018 II, 2022 II, +), plaque psoriasis (2021 III, 2020 III, 2019 III, +), psoriatic arthritis (2020), RA (2017 II), UC phase II
Subcutaneous IL 17A and IL 17F
Bivatuzumab mertansine Humanized monoclonal antibody IgG1 Antineoplastic squamous cell carcinoma, breast phase I fail x2, head/neck or esophagus phase I fail, toxicity CD44 v6
Bleselumab Human monoclonal antibody IgG4κ Disease modifying organ transplant rejection phase II 2020 to prevent FSGS in kidney transplant patients
Phase II psoriasis- medication tolerated with minimal reaction, no benefit to disease process
Intravenous CD40
Blinatumomab Blincyto
FDA 2014
EU 2015
Murine(scFv - kappa - heavy) - (scFv - heavy - kappa) BiTE Antineoplastic
Ph chrom neg pre-B ALL (CD19+) phase II
B-cellprecursor acute lymphoblastic leukemia (ALL) initial or relapsed/refractory
Intravenous Bispecific T-cell engager monoclonal antibody construct that directs CD-3 positive effector memory T cells to CD19-positive target cells
Blontuvetmab Blontress Canine monoclonal antibody IgG2 κ/λ Veterinary treat canine B-cell lymphoma CD20
Blosozumab Humanized IgG4κ Disease modifying
Osteoporosis 3 phase I and one phase 2 injection site reaction and antibodies to antibody
Intravenous
Subcutaneous
SOST
Antisclerostin
Bococizumab
RN316
PF-04950615
Humanized IgG2κ Disease modifying
Dyslipidemia
Phase III 2019
Discontinued secondary to antidrug antibodies, no primary endpoint achieved
Subcutaneous intravenous Neural apoptosis-regulated proteinase 1
PCSK9 (proprotein convertase subtilisin/kexin type 9, neural apoptosis-regulated convertase 1, NARC1, NARC-1, proproteine convertase 9, PC9)
Brazikumab Human monoclonal antibody IgG2λ Disease modifying
Ulcerative colitis phase II 2021
Phase I/II completed Crohn. Phase III ongoing
Subcutaneous IL23
Brentuximab vedotin Adcetris
FDA 2013
Breakthrough therapy status by FDA 2018
Chimeric humanized monoclonal antibody IgG1κ Antineoplastic
Hodgkinlymphoma
Anaplasticlarge-cell lymphoma
Intravenous CD30 (TNFRSF8) an antibody-drug conjugate (ADC) 3 parts: anti-CD30 (cAC10, a cell membrane protein of the tumor necrosis factor receptor), a microtubule disrupting agent monomethyl auristatin E (MMAE) and a protease-cleavable linker that attaches MMAE covalently to cAC10. The combination disrupts the intracellular microtubule network causing cell-cycle arrest and apoptotic cellular death
Briakinumab Human monoclonal antibody Disease modifying psoriasis,
Drug development stopped for psoriasis, phase IIb study in Crohn's
Intravenous IL-12, IL-23
Brodalumab
AMG827
Siliz
FDA 2016
Human monoclonal antibody IgG2κ Disease modifying
Plaque psoriasis
Completed phase III
Subcutaneous Receptor IL-17RA
Brolucizumab
RTH258
ESBA1008
FDA review 2018 Humanized single chain antibody fragment (scFv κ) Disease modifying
Wet or age-related macular degeneration phase III to be completed Sept 2018, 2020 HAWK (NCT02307682) and HARRIER (NCT02434328) phase III trials good results
Intravitreal
https://www.novartis.com/news/media-releases/new-novartis-phase-iii-data-brolucizumab-demonstrate-reliability-12-week-treatment-interval
VEGFA
Brontictuzumab Humanized IgG2λ Antineoplastic
Phase I
Colorectal
Lymphoid
Adenoid cystic
Solid tumors
Intravenous Notch 1
Burosumab
KRN23
Crysvita
FDA 2018
Human monoclonal antibody IgG1κ Disease modifying
X-linked hypophosphatemia
Phase III completed
Subcutaneous
https://www.creativebiolabs.net/burosumab-overview.htm
FGF 23 phosphaturic hormone fibroblast growth factor 23
Cabiralizumab Humanized monoclonal antibody IgG4κ Antineoplastic metastatic pancreatic cancer phase II 2020
Many other cancers phase I
Intravenous CSF1R
Camidanlumab tesirine
ADCT-21
Human monoclonal antibody Antineoplastic
B-cell Hodgkin's lymphoma, non-Hodgkin lymphoma, acute lymphoblastic leukemia, acute myeloid leukemia 2018 phase I
Advanced solid tumors with literature evidence of CD25(+) treg content
Head and neck
Nonsmall cell lung
Gastric, esophageal,
Pancreas, bladder,
Renal cell, melanoma,
Triple-negative breast, ovarian phase I 2021
Intravenous CD25
Sinilimab
Camrelizumab
IBI308
China pending approval Humanized monoclonal antibody IgG4κ Antineoplastic
Phase III nasopharyngeal cancer 2021
Phase III esophageal cancer 2021
Programmed cell death 1 (PDCD1)
Canakinumab
ACZ885
Ilaris
FDA 2009
EU 2009
Human monoclonal antibody IgG1κ Disease modifying
Cryopyrin-associatedperiodic syndromes
Includingfamilial cold auto-inflammatory syndrome andMuckle–Wells syndrome; tumor necrosis factor receptor-associated periodic syndrome (TRAPS); hyperimmunoglobulin Dsyndrome(HIDS)/mevalonate kinase deficiency (MKD) and familial Mediterranean fever (FMF)
SystemicJuvenile idiopathic arthritis
Treat Juvenile idiopathic arthritis phase III
NSCLC 2025 phase III
CVD rejected by FDA
Behcet
Subcutaneous IL-1β
Cantuzumab mertansine Humanized monoclonal antibody IgG1κ Antineoplastic
Colorectal cancer phase I 2007
Intravenous Mucin CanAg
Cantuzumab ravtansine Humanized monoclonal antibody IgG1κ Antineoplastic
Cancers
MUC1
Caplacizumab-yhdp Cablivi
(Nanobody program)
FDA 2019
EU 2018
Humanized single variable domain antibody (bivalent nanobody) Disease modifying
Inhibits interaction vWF and platelets
Treat acquired TTP
Phase III Hercules study completed
Intravenous
Subcutaneous
VWF
Capromab pendetide Prostascint
FDA 1996
Murine monoclonal antibody Diagnostic imaging
Prostatic carcinoma cells detection
Intravenous Tumor surface antigen PSMA
Carlumab Human monoclonal antibody IgG1κ Antineoplastic
Prostate phase II no long term benefit
Pulm fibrosis phase II no benefit
Intravenous hMCAF/MCP-1 (human macrophage/monocyte chemotactic protein-1)
Carotuximab
TRC105
Chimeric monoclonal antibody IgG1κ Antineoplastic angiosarcoma
Hepatocellular car phase I/II 2020
Glioblastoma multi-phase II 2014 terminated poor accrual ?results
Angiosarcoma phase III 2019 TAPPAS trial
Prostate ca phase II 2021
NSCLC phase I 2019
Intravenous Endoglin (CD105)
Catumaxomab Removab
FDA approved pend 2017)
EU approved 2009
Removab: A trifunctional rat/murine hybrid antibody IgG2a/IgG2b Antineoplastic
Removab
Ovarian cancer phase II, malignant ascites phase II, gastric cancer phase II (ovarian, gastric, colon, pancreatic, breast, endometrial)
Proxinium
Head and neck cancer
Intraperitoneal EpCAM, CD3
Catumaxomab consists of one “half” (one heavy chain and one light chain) of an anti-EpCAM antibody and one half of an anti-CD3 antibody, so that each molecule of catumaxomab can bind both EpCAM and CD3. In addition, the Fc-region can bind to an Fc receptor on accessory cells like other antibodies, which has led to calling the drug a trifunctional antibody.
cBR96-doxorubicin immunoconjugate
aka SGN-15
Humanized monoclonal antibody IgG1κ Antineoplastic
Cancer
Sponsorship ceased 2005
Cedelizumab CIMZIA Humanized monoclonal antibody IgG4κ Prevent organ transplant rejection CD4
Cemiplimab Libtayo
FDA 2018
Human monoclonal antibody IgG4 Antineoplastic
Nonsmall cell lung cancer (NSCLC) phase I 2021, phase III 2022 × 3
Oropharynx phase II 2022
Multiple myeloma phase II 2022
Ovarian ca phase II 2022
Head neck squamous cell carcinoma phase II 2020
Cutaneoussquamous cell
Glioblastoma multiforme phase II 2021
Lung ca phase II 2022
Cervical cancer phase III 2023
Intravenous Programmed cell death receptor PCDC1
Cergutuzumab amunaleukin
Aka RO6895882, CEA-IL2v
Humanized monoclonal antibody Antineoplastic phase I Dec 2018 Intravenous IL2
Certolizumab pegol
CDP870
Cimzia
FDA 2008
EU 2009
Recombinant, humanized antibody Fab’ fragment Disease-Modifying
Crohns
Rheumatoidarthritis (phase IIIs completed)
Psoriatic arthritis phase III
Ankylosing spondylitis
Subcutaneous Tumor necrosis factor α blocker
Cetrelimab Relatimab Human monoclonal antibody IgG4κ Antineoplastic Nothing on PubMed or creative lab
Substance is registered with FDA
Programmed cell death 1
Cetuximab
IMC-225
Leukeran
Erbitux
FDA 2004
EU 2004
Recombinant chimeric monoclonal antibody IgG1κ Antineoplastic agent
Metastaticcolorectal cancer and head and neck cancer
NSCLC
Intravenous solution EGFR
Citatuzumab bogatox Humanized Fab IgG1κ Antineoplastic ovarian cancer and other solid tumors Study phase I terminated 2008 EpCAM
Cixutumumab Human monoclonal antibody IgG1κ Antineoplastic
Solid tumors
Sarcoma phase II
Esophageal cancer phase II
Rhabdomyosarcoma phase II no benefit
Liver cancer phase I
Low antitumor effect
Pancreas no benefit 2012
Intravenous IGF-1 receptor (CD221)
Clazakizumab
ALD−518
Humanized monoclonal antibody Disease modifying rheumatoid arthritis phase II 2015 × 3
Crohn disease phase II 2013
Highly sensitized renal transplant candidates phase II 2020
Treat post-tx rejection kidney phase II 2020
Antibody-mediated rejection phase III 2027
Subcutaneous IL6
Clenoliximab Chimeric monoclonal antibody Disease modifying Rheum Arth
No study since 2003
CD4
Clivatuzumab tetraxetan
(90)Y-clivatuzumab tetraxetan
hPAM4-Cide Humanized monoclonal antibody IgG1κ Antineoplastic
Pancreatic cancer
Phase III 2017 PANCRIT-1 study. Study terminated no increase improvement of overall survival
MUC1
Codrituzumab Humanized monoclonal antibody IgG1κ Antineoplastic
HCC
Phase Ib no response
Phase II no response
Glypican 3
Cofetuzumab pelidotin Humanized monoclonal antibody IgG1κ Antineoplastic Nothing on PubMed or creative lab
Substance is not registered with FDA
Protein tyrosine kinase 7 (PTK7)
Coltuximab ravtansine
SAR3419
Chimeric monoclonal antibody IgG1 conjugated to DM4 (N2′-(4-((3-carboxypropyl)dithio)-4-methyl-1-oxopentyl)-N2′-deacetylmaytansine) Antineoplastic
Relapse/refractory ALL phase II 2015 low clinical response
Phase II moderate response
CD19
Conatumumab
AMG655
Human monoclonal antibody IgG1κ Antineoplastic
Phase II 2019: Advanced solid tumors
Carcinoid
Colorectal cancer
Locally advanced
Lymphoma
Metastatic cancer
Nonsmall cell lung cancer
Sarcoma
Solid tumors
Colon cancer phase Ib/II no benefit
Intravenous TRAIL-R2
Concizumab Humanized IgG4κ Disease modifying
Hemophilia A and B phase II 2020
Subcutaneous Kunitz-type protease inhibitor 2 domain of tissue factor pathway inhibitor (TFPI)
Cosfroviximab ZMapp Chimeric monoclonal antibody IgG1κ
Triple monoclonal antibody cocktail
Disease modifying Ebola virus
Ongoing studies show benefit but not enough enrolled to power study
Ebola virus glycoprotein
Crenezumab
RG7412
MABT5102A
Humanized monoclonal antibody IgG4 Disease modifying
Alzheimer's disease phase III study ongoing prodromal/mild AD 2021
Phase III 2022
Intravenous 1-40-β-amyloid
Crizanlizumab
SelG1
FDA review possible 2019 Humanized monoclonal antibody IgG2κ Disease modifying
Sickle cell disease phase II 2022 children
Phase II adults decrease pain crisis
Intravenous P Selectin
Crotedumab Human monoclonal antibody IgG4κ Disease modifying DM type II No results GCGR
Cusatuzumab
ARGX-110
Humanized monoclonal antibody IgG1 Antineoplastic
Phase I completed safe
Phase I/II CTCL dec 2018
Nasopharyngeal carcinoma 2018
Intravenous CD70
Dacetuzumab
HU-S2C6
ASKP1240
SGN-40
Humanized monoclonal antibody IgG1 Antineoplastic
Hematologic cancers
Multiple myeloma phase I 2007
Large B-cell lymphoma phase II 2009 enrollment stopped no benefit
CLL phase II 2006
NHL phase I
Renal transplant (CIRRUS I) phase II 2022
SLE nephritis phase II 2020
Intravenous CD40
Daclizumab Zenapax
Zinbryta
FDA 1997
EU 1999
Zenapax withdrawn from market Apr 2009 for commercial reasons
Zinbryta withdrawal 2018 secondary to risk/benefit profile
Humanized monoclonal antibody IgG1κ Disease modifying
Prevention of organ transplant rejections
Phase IV kidney transplants, multiple sclerosis phase III 2018 pulled from market secondary inflammatory brain disorders Biogen
Heart transplant phase IV 108 studies Zanapax discontinued from market by Roche (basiliximab replace)
CD25 (α chain of IL-2 receptor)
Dalotuzumab Humanized monoclonal antibody IgG1κ Antineoplastic
Phase I multiple
Phase II breast no improvement × 2
Phase III colon no improvement
Ped solid phase I
Intravenous IGF-1 receptor (CD221)
Dapirolizumab pegol Humanized monoclonal antibody IgG1κ SLE phase II Nov 2018
Phase I safe
Intravenous CD154 (CD40L)
Daratumumab Darzalex
FDA 2015
EU 2016
Human IgG1κ Antineoplastic agent
Multiple myeloma relapse/refractory
Phase III completed
Intravenous solution CD38
Induces CDC, ADCC, ADCP, and apoptosis
Dectrekumab
QAX576
Human monoclonal antibody IgG1κ Cancers, asthma phase II, idiopathic pulmonary fibrosis, eosinophilic Esophagitis phase II some benefit but primary endpoint not achieved, Keloids, Crohn's disease phase II trials 2013 Nothing on PubMed
Substance is registered with FDA, creative lab
IL-13
Demcizumab Humanized monoclonal antibody IgG2κ Antineoplastic
NSCLC phase II 2018
Phase I safety established with 50% tumor regression response
Intravenous Delta-like ligand 4DLL4
DLL4 and Notch1, signaling stimulated by DLL4 plays a role in development of blood vessels throughout life
Denintuzumab mafodotin
HBU-12
SGN-CD19A
Humanized monoclonal antibody IgG1κ
Antibody-drug conjugate (ADC) composed of a humanized anti-CD19 monoclonal antibody conjugated to the microtubule-disrupting agent monomethyl auristatin F (MMAF)
Antineoplastic
LBCL phase II terminated study by company
Acute lymphoblastic leukemia and B-cell non-Hodgkin lymphoma
Phase I 2017
Phase II 2018 terminated by sponsor
Intravenous CD19
Denosumab
AMG162
Prolia
FDA 2010
EU 2010
Xgeva
FDA 2011
EU 2011
Human monoclonal antibody IgG2 Disease modifying
Osteoporosis FREEDOM trial, bone metastases, etc. 186 studies
Phase III completed
Melanoma phase II 2022
Bone giant cell tumor phase II 2025
Subcutaneous Receptor activator of nuclear factor kappa-B ligand (RANKL)
Xgeva: Prevention of skeletal-related events (SREs) in adults with bone metastases from breast and castration-resistant prostate cancer.
Prolia: Osteoporosis
Depatuxizumab mafodotin
ABT 414
Chimeric humanized monoclonal antibody IgG1κ CONJUGATED TO AURISTATIN F Glioblastoma
Phase III Nov 2019
Children phase III 2020
Intravenous EGFR
Derlotuximab biotin
Iodine (131 I) derlotuximab biotin
Chimeric monoclonal antibody IgG1κ Immunoassays
Potential for glioblastoma multiforme
Histone complex
Detumomab Murine monoclonal antibody IgG1 Antineoplastic
B-lymphoma cell
Nothing on PubMed or clinical trials
Substance is not registered with FDA, is on creative lab
CD3E
Dezamizumab
GSK-2398852
Humanized monoclonal antibody IgG1κ Disease modifying
Treat amyloidosis
Transthyretin cardiomyopathy amyloidosis (ATTR-CM), suspended pending data review Aug 2018 phase I x 4
Intravenous Serum amyloid P component
Dinutuximab
APN311
Unituxin
FDA 2015
EU 2015 then withdrawn EU
Chimeric monoclonal antibody IgG1κ Antineoplastic
Neuroblastoma phase I 2022
SCLC phase III Nov 2019
Osteosarcoma phase II Dec 2018
Neuroblastoma phase II 2020
Intravenous GD2 ganglioside
Diridavumab
CR6261
Human monoclonal antibody IgG1λ Disease modifying
Infectious disease/influenza A
Very good response in animal study mice
Phase II 2019
Intravenous Influenza A hemagglutinin
Domagrozumab
PF-06252616
Humanized monoclonal antibody IgG1κ Disease modifying
Duchenne muscular dystrophy phase II 2018
Phase I completed
GDF-8
Dorlimomab aritox F(ab')2 Murine Nothing on PubMed or clinical trials
Substance is not registered with FDA, or creative lab
Dostarlimab
TSR042
WBP285
FDA review pending 2019 Humanized monoclonal antibody IgG4κ Antineoplastic
Solid tumor
Phase I, II, III studies ongoing
Ovarian CA (first study) phase III 2023
Programmed cell death protein-1 (CD279) PCDP1
Drozitumab
PRO95780
rhuMAB DR5
Human monoclonal antibody IgG1λ Antineoplastic
Colorectal cancer Ib 2012
Preclinical rhabdomyosarcoma 2018
Chondrosarcoma not efficacious
NHL results?
Intravenous Death receptor 5 (DR5)
Duligotuzumab
MEHD7945A
Human monoclonal antibody IgG1κ Antineoplastic squamous head and neck phase II no benefit
Colon ca phase II no benefit
Anti-EGFR × Anti-HER3 bispecific antibody
Dupilumab Dupixent
FDA 2017
Human monoclonal antibody IgG4 Disease modifying asthma, atopic dermatitis
Ongoing studies
Subcutaneous IL4
Durvalumab Imfinzi
FDA 2017
Human monoclonal antibody IgG1κ Antineoplastic agent
Treat NSCLC stage III phase I, urothelial carcinoma
Intravenous PD-L1 (CD274) and CD80—inhibit binding of programmed death ligand 1 to PD-1 and CD80 allowing T cell to recognize and kill tumor cells
Dusigitumab
MEDI 573
Human monoclonal antibody IgG2λ Antineoplastic
Breast cancer phase II results?
HCC phase II results?
Intravenous ILGF2
Duvortuxizumab
MGD011
Chimeric/humanized monoclonal antibody Antineoplastic
B-cell malignancy
Phase I/II Jul 2018/2020
Intravenous CD19, CD3E
Ecromeximab
KW2871
Chimeric monoclonal antibody IgG1κ Antineoplastic
Metastatic melanoma
Phase I/II clinical activity limited
Intravenous GD3 ganglioside
Eculizumab Soliris
PNH
FDA 2007
EU 2007
aHUS, and myasthenia gravis
FDA 2018
EU 2018
Japan 2018
Humanized monoclonal antibody IgG1/4 Immuno-regulation
Paroxysmalnocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (HUS)
Generalizedmyasthenia gravis (MG)
Phase II CAD
Intravenous C5
Edobacomab
E5
Murine monoclonal antibody No improved survival Endotoxin
Edrecolomab Panorex Murine monoclonal antibody IgG2κ Antineoplastic
Colorectal carcinoma phase III 2003 no improvement
Intravenous Glycoprotein EpCAM/17-1A
Efalizumab Raptiva
FDA 2003
EU 2004
Withdrawn both markets 2009
Recombinant humanized monoclonal antibody IgG1κ Disease modifying
(2003 approved) psoriasis
Subcutaneous
Voluntary withdrawal 2009
Human CD11a
Increase risk progressive multifocal leukoencephalopathy (PML)
Efungumab
MYC123
Mycograb
Mycograb C28Y
Human scFv Antiinfectious agent
Invasive Candida infection
Intravenous Heat shock protein 90 (Hsp90)
Eldelumab
Mdx 1100
Human monoclonal antibody IgG1κ Crohn's disease phase IIa no significant response, ulcerative colitis phase IIb prim endpoint not achieved
Rheum arthritis phase II
Intravenous Interferon γ-induced protein
CXCL 10
Elezanumab
PR-1432051
ABT-555
Human monoclonal antibody IgG1λ Spinal cord injury and multiple sclerosis phase II 2021 Intravenous REPULSIVE GUIDANCE MOLECULE FAMILY MEMBER A (RGMA)
Elgemtumab
LJM716
Human IgG1κ Antineoplastic
Breast gastric phase I
Intravenous ERBB3 (HER3)
Elotuzumab
PDL063
Empliciti
FDA 2015
EU 2016
Human IgG1κ Antineoplastic
Multiple myeloma
Phase III completed and ongoing
Intravenous SLAMF7
Elsilimomab
B-E8
Humanized monoclonal antibody IgG1 Antineoplastic multiple myeloma
Not effective in mice
IL-6
Emactuzumab
RG7155
Humanized monoclonal antibody IgG1 ANTINEOPLASTIC
Phase I 2019 solid tumors
Phase II 2025 REDIRECT study ovarian, fallopian tube cancer
Pancreatic phase II 2020
HUMAN MACROPHAGE COLONY-STIMULATING FACTOR RECEPTOR (CSF1R, CD115)
Emapalumab
NI-0501
Gamifant
FDA 2018
EU pending
Human monoclonal antibody IgG1λ Hemophagocytic lymphohistiocytosis
Phase III 2021
Intravenous Interferon γ
Emibetuzumab
LA480
LY2875358
Humanized monoclonal antibody IgG4κ
Bivalent antibody
Antineoplastic
NSCLC phase II 2020
Advanced cancer
Gastric safe ?effective adenocarcinoma
Gastroesophageal junction adenocarcinoma
Hepatocellular cancer
Renal cell carcinoma
Nonsmall cell lung cancer phase II Jan 2018
Phase I safe with tumor response
Intravenous Hepatocyte growth factor receptor (HHGFR) and MET signaling
Emicizumab
ACE910
Hemlibra
FDA 2018
Humanized monoclonal antibody IgG4κ
Bispecific
Disease modifying
Hemophilia A phase III 2020
With or without inhibitors
Subcutaneous Activated F9, F10
Enapotamab vedotin Human monoclonal antibody IgG1κ Antineoplastic Nothing on PubMed or clinical trials
Substance is not registered with FDA, or creative lab
Human growth factor receptor AXL
Enavatuzumab
PDL192
Humanized monoclonal antibody IgG1κ Antineoplastic
Phase I 2011
No responses and liver pancreatic toxicity
Intravenous TWEAK receptor
Enfortumab vedotin FDA review pending 2019 Human monoclonal antibody Antineoplastic bladder cancer phase I
Phase II ongoing
Nectin-4
Anti-Nectin-4
Monoclonal antibody attached to a microtubule-disrupting agent, monomethyl auristatin E (MMAE)
Enlimomab pegol Murine monoclonal antibodyIgG2a Disease modifying
Stroke
Nothing on PubMed or clinical trials
Substance is not registered with FDA
ICAM-1 (CD54)
Enoblituzumab
MGA 271
Humanized monoclonal antibody IgG1κ Antineoplastic
Phase I 2022 children Neuroblastoma
Rhabdomyosarcoma
Osteosarcoma
Ewing sarcoma
Wilms tumor
Desmoplastic small round cell tumor
Phase I melanoma, NSCLC 2018
Phase II prostate 2021
CD276 (B7–H3)
Enokizumab
MEDI528
Humanized monoclonal antibody IgG1κ Asthma phase II
No improvement
Intravenous IL9
Enoticumab
REGN421
Human monoclonal antibody IgG1κ Antineoplastic
Phase 1 2014 ovarian cancer +
Delta-like canonical notch ligand 4 (DLL4)
Ensituximab
NEO-201
NPC-1C
Chimeric monoclonal antibody IgG1κ Antineoplastic
Phase II pancreatic and colorectal cancer 2017
Intravenous 5AC
Enterecept
RHU-TNFR:FC
Enbril
FDA 2003
1-235-Tumor necrosis factor receptor fusion protein attached to recombinant human IgG1 Fc fragment Disease modifying
Antirheumatic drug
Not effective for inflammatory bowel disease
Subcutaneous TNFα
Epitumomab cituxetan
AS-1402
HuHMFG-1
Sontuzumab Humanized monoclonal antibody IgG1 Antineoplastic
Breast cancer phase II 2012 no benefit
Episialin
MS4A1 (membrane-spanning 4-domains subfamily A member 1, CD20 (HMFG-1)
Epratuzumab
HLL2
AMG412
Humanized monoclonal antibody IgG1κ
ADCC/CDC
Antineoplastic
B-ALL phase III ongoing 2018
Disease modifying
SLE phase III no improvement
Intravenous CD22
Eptinezumab
ALD403
FDA review possible 2019 Monoclonal antibody IgG1κ Disease modifying
Migraine phase III
Calcitonin gene-related peptide
Erenumab Aimovig
FDA May 2018
Human monoclonal antibody IgG2λ Disease modifying
Migraine phase III
Calcitonin gene-related peptide (CGRP)
Erlizumab
Rhumab CD18
Humanized IgG1 F(ab')2 fragment Antineoplastic
(lab tests)
Immunosuppressive drug phase I study cough up blood and phase II did not meet goals
Heart attack, stroke, traumatic shock ??no successful CD18 drug to date
LGL type leukemia ITGB2 (CD18) and LFA-1 block growth factor of blood vessels stop lymphocytes from moving into inflamed tissue
Ertumaxomab Rexomun Rat/murine hybrid triomab, murine IgG2a HET2 target, RAT IgG2bλ CD3 target Antineoplastic
Breast
Gastric, esophageal
Phase II studies terminated company to focus on other plans not safety concerns concentrate on catumaxomab
Phase I found safe 2016
Intravenous HER2/neu, CD3
Etaracizumab or etaratuzumab
MEDI-522
Abegrin
Vitaxin
Humanized monoclonal antibody IgG2κ Antineoplastic
Melanoma phase II 2010 not beneficial, prostate cancer, ovarian cancer small and large bowel cancer phase I and II completed results unreported 2017
Intravenous Integrin αvβ3
Etigilimab Humanized monoclonal antibody IgG1κ Nothing on pubmed or clinical trials
Substance is registered with FDA, or is not in creative lab
TIGIT T-cell immunoreceptor with Ig and ITIM domains
Etrolizumab
PRO145223
RHUMAB BETA7
Humanized monoclonal antibody IgG1κ Disease modifying
Inflammatory bowel disease UC phase III 2020 × 4/2023/2024/2025
Crohn phase III 2021
Subcutaneous Integrin β7
Inhibits binding of
αEβ7 to E-cadherin
Evinacumab
REGN1500
Human monoclonal antibody IgG4κ Disease modifying
Dyslipidemia
Phase II 2020
Phase III 2020/2022
Angiopoietin 3
Evolocumab Repatha
FDA 2015
EU 2015
Human monoclonal antibody IgG2λ Disease modifying hypercholesterolemia
Completed phase III
Heterozygous familial hypercholesterolemia, CVD
Subcutaneous Proprotein convertase subtilisin kexin type 9 (PCSK9)
Exbivirumab Humanized monoclonal antibody IgG1λ Disease modifying prevent disease Hep B Oral therapy
Abstract of randomized study of 50 patients
Hepatitis B surface antigen
Fanolesomab
RB5-IGM
NeutroSpec Murine monoclonal antibody Diagnostic imaging
Appendicitis (diagnosis only)
CD15
Faralimomab Murine monoclonal antibody IgG1 Nothing on PubMed or clinical trials
Substance is not registered with FDA, or is not in creative lab
Interferon receptor
Faricimab
RG7716
RO6867461
Humanized monoclonal antibody IgG1mab Disease modifying angiogenesis, ocular vascular diseases
STAIRWAY, BOULEVARD, RHINE, Yosemite phase II and III studies phase III 2022 for diabetes maculae edema
AMD LUCERNE phase III 2022 TENAYA phase III 2022
Intravitreous ANTIVASCULAR ENDOTHELIAL GROWTH FACTOR/ANTIANGIOPOIETIN 2 BISPECIFIC ANTIBODY (VEGF-A and Ang-2)
Farletuzumab
MORAB-003
Humanized monoclonal antibody IgG1κ Antineoplastic
Ovarian cancer phase III subgroup may benefit
Intravenous Folate receptor 1
Fasinumab
REGN475
SAR164877
MT5547
Human monoclonal antibody IgG4κ Disease modifying acute sciatic pain phase III
Knee arthritis pain phase III 2021
Subcutaneous (auto injector) Human nerve growth factor (HNGF)
FBTA05
Bi20
Lymphomun Rat IgG2b (CD3)/murine IgG2a (CD20) hybrid trifunct Antineoplastic
Chronic lymphocytic leukemia trial terminated recruitment too slow
Intravenous? CD20/CD3
Felvizumab Humanized monoclonal antibody IgG1κ Antiinfectious agent
Respiratory syncytial virus infection
Nothing on PubMed or clinical trials
Substance is not registered with FDA, but is in creative lab
Respiratory syncytial virus
Fezakinumab Human monoclonal antibody IgG1λ Disease modifying
Rheumatoid arthritis, psoriasis (not good for)
Atopic dermatitis phase IIb good results
Intravenous IL-22
Fibatuzumab
Ifabotuzumab
Humanized monoclonal antibody IgG1κ Disease modifying
Myelodysplastic syndrome
Research in Australia for GBM phase I
Ephrin receptor A3
Ficlatuzumab
SCH 900105
AV 299
Humanized monoclonal antibody IgG1κ Antineoplastic
Head and neck cancer phase I 2020
Pancreatic phase I 2023
NSCLC phase I/II 2013
AML phase I 2020
Intravenous Hepatocyte growth factor (HGF) hepapoietin A
Figitumumab
CP751871
Human monoclonal antibody IgG2κ
Ceased development in 2011 by pfizer
Antineoplastic
Adrenocortical carcinoma, nonsmall cell lung carcinoma etc. ?additional benefit in phase I
Insulin-like growth factor receptor IGF-1 receptor (CD221)
Firivumab Human monoclonal antibody IgG1κ Disease modifying
Influenza A virus hemagglutinin
Nothing on PubMed or clinical trials
Substance is registered with FDA, and is in creative lab
INFLUENZA A VIRUS HEMAGGLUTININ HA
Flanvotumab
IMC20D7S
Human monoclonal antibody IgG1κ Antineoplastic
Melanoma phase I 2012
Intravenous
No published data
TYRP1 (glycoprotein 75)
Fletikumab Human monoclonal antibody IgG4 Disease modifying
Rheumatoid arthritis
phase IIa good, phase IIb no results
IL 20
Flotetuzumab
MGD006
S80880
Humanized di-scFv
dual affinity retargeting (DART) to CD123 and CD3
Antineoplastic
Hematologic malignancies (ALL, NHL)
Phase II not yet recruiting 2018
Intravenous? IL 3 receptor
Fontolizumab HuZAF Humanized monoclonal antibody IgG Disease modifying
Treat Crohn's clinical development stopped despite some benefit phase II ustekinumab is better
IFN-γ
FOR46 Antibody drug conjugate Antineoplastic
Phase I for multiple myeloma failed remission or relapse
Phase I prostate cancer
Intravenous CD46
Foralumab Human monoclonal antibody IgG1κ Disease modifying NASH phase II 2019 Oral CD3 epsilon
Foravirumab Human monoclonal antibody IgG1κ Disease modifying rabies (prophylaxis) Nothing in pub med or clinical trials Rabies virus glycoprotein
Fremanezumab
LBR-101
RN307
Ajovy
FDA 2018
Humanized monoclonal antibody IgG2κ Disease modifying migraine and cluster headache phase III 2019 Subcutaneous α-Calcitonin gene-related peptide
Fresolimumab Humanized monoclonal antibody IgG4 Disease modifying
Idiopathic pulmonary fibrosis (IPF), scleroderma, focal segmental glomerulosclerosis (phase 2), cancer (kidney cancer and melanoma)
Need larger study for FSGS
Good response scleroderma
https://newdrugapprovals.org/2016/01/30/fresolimumab/
TGF β 1
Frovocimab
LY3015014
Humanized monoclonal antibody IgG4κ Disease modifying hypercholesterolemia
Completed phase II trials 2014 good response and safe
Subcutaneous PROPROTEIN CONVERTASE SUBTILISIN KEXIN 9 (PCSK9)
Frunevetmab https://en.wikipedia.org/wiki/List_of_therapeutic_monoclonal_antibodies - cite_note-WHOList 116-17
NV-02
Veterinary monoclonal antibody IgG1κ Veterinary Feline muscle nerve growth factor
Fulranumab
AMG403
Human monoclonal antibody IgG2κ Disease modifying
Pain osteoarthritis pain phase III 2017
Subcutaneous Nerve growth factor
Galcanezumab
LY2951742
Emgality
FDA 2018
Humanized monoclonal antibody IgG4κ Disease modifying
Migraine
Phase III completed
Cluster HA phase III 2020
Subcutaneous Calcitonin gene-related polypeptides (CGRPs) α and β
Galiximab
IDEC-114
Chimeric monoclonal antibody IgG1λ
ADCC/CDC
Antineoplastic lymphoma phase II 2015 minimal response ORR 10.3% Intravenous CD80
Gancotamab
MM-302
Human cFv
Single chain fragment
Antineoplastic
Breast cancer phase I–III
Intravenous HER2/neu
Ganitumab
AMG479
Human monoclonal antibody IgG1κ Antineoplastic
Pancreatic phase III—no increase benefit
Phase III for rhabdomyosarcoma and Ewings 2021
Not beneficial in NSCLC
Intravenous IGF-1 receptor (CD221)
Gantenerumab
R04909832
R1450
Human monoclonal antibody IgG1κ Disease modifying
Alzheimers
Phase III stopped for potential futility additional studies at higher dosing (DIAN in a phase II/III trial in individuals at risk
For and with early-stage autosomal-dominant AD phase III 2021
Subcutaneous Beta amyloid
Gatipotuzumab PankoMab-GEX Humanized monoclonal antibody IgG1κ Antineoplastic
Ovarian, non-small cell lung cancer (NSCLC), colorectal cancer (CRC), breast cancer (BC), gynecological cancers (GYN) phase I used for diag/prog now
Intravenous Musculus, antimucin (MUC1)
Gavilimomab
ABX-CBL
Murine monoclonal antibody IgM Disease modifying
Graft versus host disease phase III completed 2005 less effective than antithymocyte antibody
CD147 (basigin)
Gedivumab
RG7745
RO6876802
Human monoclonal antibody IgG1κ Disease modifying
Influenza virus A
No studies
PubMed/clinical trials
Influenza virus hemagglutinin HA
Gemtuzumab ozogamicin Mylotarg
AML FDA 2000
Voluntary withdrawal 2010 VOD now black box warning
Returned to market with FDA approval 2017
Humanized monoclonal antibody IgG4/toxin conjugate Antineoplastic
Acute myelogenous leukemia
Many ongoing and completed studies
Intravenous CD33
Gevokizumab
XOMA 052
Humanized monoclonal antibody IgG2κ Disease modifying DM phase II (late stage) no results
Other dz too 24 studies behcet uveitis failed primary end point phase III (Eyeguard _B) 2015
Subcutaneous IL-1β
Gilvetmab
PD1
Veterinary monoclonal antibody IgG2κ Antineoplastic No studies clinical trial, pub med CANIS FAMILIARIS PROGRAMMED CELL DEATH PROTEIN 1 (PCDC1)
Gimsilumab
MORAb-022
Human monoclonal antibody IgG1 Disease modifying
Rheumatoid arthritis
Asthma
Phase I poster presentation of safety results good 2016
Intravenous HUMAN GRANULOCYTE-MACROPHAGE COLONY-STIMULATING FACTOR (CSF2)
Girentuximab
WX-G250
CG250
Rencarex
Reductane
Chimeric monoclonal antibodyIgG1κ
Radioactive labeled ab
Antineoplastic
Clear cell renal cell carcinoma for treatment and imaging
Intravenous Carbonic anhydrase 9 (CA-IX)
Glembatumumab vedotin
CR011
Human monoclonal antibody IgG2κ
Antibody drug complex
Antineoplastic
Melanoma phase II, breast cancer
Intravenous Human glycoprotein NMB extracellular domain (GPNMB)
Golimumab
CNTO 148
Simponi
FDA 2009
EU 2009
Human monoclonal antibody IgG1κ Disease modifying
Rheumatoidarthritis, psoriatic arthritis, juvenile rheum arth, ankylosing spondylitis many studies, UC, DM1 phase I (2020, 2021)
Subcutaneous, intravenous TNF-α
Gomiliximab
IDEC-152
Lumiliximab
ST-152
Chimeric monoclonal antibody IgG1κ
ADCC/CDC
Allergic asthma ?
Antineoplastic CLL
Phase I, phase 2/3 2014
Failed efficacy
CD23 (IgE receptor)
Gosuranemab
BIIB092
IPN-007
FDA orphan drug status Humanized monoclonal antibody IgG4κ Progressive supranuclear palsy
Phase I 2020
Alzheimer 2021
Intravenous τ protein
Guselkumab
CNTO 1959
Tremfya
FDA ?
Human monoclonal antibody IgG1λ Disease modifying
Psoriasis
Adenomatous polyposis
Subcutaneous IL23A
Hu3F8 Humanized monoclonal antibody IgG3 Antineoplastic
Phase I
For neuroblastoma mod tox and substantial effect on tumor
Phase II ongoing
Intravenous GD2 ganglioside
Ianalumab Human monoclonal antibody IgG1κ Immunomodulation
Autoimmune hepatitis
Human cytokine receptor BAFF-R
Ibalizumab Trogarzo
FDA/EU approved
Humanized monoclonal antibody IgG4 Disease modifying anti-HIV
Phase III
CD4
Ibritumomab tiuxetan
IDEC-129
IDEC-IN2B8
IDEC-Y2B8
Zevalin
FDA 2002
EU 2004
Murine monoclonal antibody IgG1κ YT77 or In98 bound Antineoplastic
Follicularnon-Hodgkin's lymphoma, B-cell NHL, multiple myeloma conditioning for BMT, B-cell DLCL, mantle cell
Many studies
CD20 (human B-lymphocyte-restricted differentiation antigen, Bp35)
Icrucumab
IMC 18F1
Human monoclonal antibody IgG1κ Antineoplastic
No benefit breast phase II
No benefit colon phase II
No benefit urothelial phase II
Intravenous Vascular endothelial growth factor receptor (VEGFR-1)
Idarucizumab Praxbind
FDA 2015
EU 2015
Humanized monoclonal antibody Fab fragment Antidotes
Drug reversal agent
Reversal of anticoagulant effects of dabigatran
Phase III trial RE-VERSE AD
Intravenous Dabigatran etexilate
Igovomab Indimacis-125 Murine F(ab')2 Diagnostic imaging
Ovarian cancer (diagnosis)
Iladatuzumab vedotin
RG7986
Humanized monoclonal antibody IgG1κ Antineoplastic Nothing in PubMed or clinical trials Human gene B29 protein (CD97B)
Imalumab
BAX69
Human monoclonal antibody IgG1κ Antineoplastic Intraperitoneal infusion, intravenous Macrophage migration inhibitory factor (MIF)
Imaprelimab Humanized IgG1κ Antineoplastic Nothing in PubMed or clinical trials Melanoma cell adhesion molecule (MCAM)
Imciromab pentetate Myoscint
FDA approved 1996
Withdrawn from market
Murine monoclonal antibody fragment Fab IgG2aκ Diagnostic
Cardiac imaging
Cardiac myosin
Imgatuzumab
RG7160
RO5083945
GA201
HUMA-B
Humanized monoclonal antibody IgG1κ Antineoplastic
Colorectal 2013
Head and neck 2017
NSCLC 2017
Epidermal growth factor receptor (EGFR, HER1)
IMGN632 Monoclonal antibody with antibody drug conjugate Antineoplastic
AML, ALL phase I 2021
NCT03386513
Intravenous CD123
Inclacumab
RG1512
RO4905417
Human monoclonal antibody IgG4κ Disease modifying
Cardiovascular disease phase II
Intravenous Selectin P
Indatuiximab ravtansine Chimeric monoclonal antibody IgG4κ Antineoplastic
Preclinical breast cancer
CD138 (syndecan-1) SDC1
Indusatumab vedotin
TAK-264
MLN0264
Human monoclonal antibody IgG1κ conjuagated via a mc-val-cit-PABC linker to monomethyl auristatin E {MMAE (5F9-mc-val-cit-PABC-MMAE)} Antineoplastic
Gastrointestinal, pancreatic, gastroesophageal
Safe phase I, phase II pancreatic min response, three studies terminated by company business
Intravenous Guanylate cyclase C (GUCY2C)
Inebilizumab
MEDI-551
Humanized monoclonal antibody IgG2κ ADCC Antineoplastic
Refractory DLBCL phase II 2016
Disease modifying systemic sclerosis, multiple sclerosis
Neuromyelitis optica
Intravenous CD19
Infliximab Remicade
FDA 1998
EU 1999
Inflectra
FDA 2016
EU 2013
Remsima
EU 2013
Human-murine chimera IgG1κ
Human constant, murine variable region
Disease modifying
Remicade
Crohn's disease; ulcerative colitis; rheumatoid arthritis; ankylosing spondylitis; psoriatic arthrits; plaque psoriasis Inflectra
Spondylitis; ankylosing; arthritis; rheumatoid colitis; ulcerative arthritis; psoriatic Crohn's disease; psoriasis
Remsima
Spondylitis; ankylosing arthritis; rheumatoid colitis; ulcerative Crohn's disease; arthritis; psoriatic psoriasis
Intravenous solution TNF-α
Inolimomab Murine monoclonal antibody Disease modifying GVHD phase III
No better than ATG in 3 studies
Abandoned not in 2017 Cochrane review
CD25 (α chain of IL-2 receptor)
Inotuzumab ozogamicin
G544
Besponsa
FDA 2017
Humanized monoclonal antibody IgG4κ ADCC/CDC Antineoplastic
ALL phase II 2023
Multiple other studies
Intravenous CD22
Intetumumab
CNTO095
Human monoclonal antibody IgG1κ Antineoplastic
Solid tumors (prostate cancer, melanoma)
Melanoma phase II possible benefit 2011
Prostate cancer no additional benefit 2013 phase II
Intravenous CD51
Ipilimumab Yervoy
FDA 2011
EU 2011
Human monoclonal antibody IgG1κ Antineoplastic agent
Bladder carcinoma (trials ongoing)
Melanoma
Ipilimumab
MDX010
MDX101
BMS-734016
Yervoy
FDA 2011 melanoma
Metastatic renal cancer/colorectal cancer 2018
Human monoclonal antibody IgG1κ Antineoplastic
Melanoma(checkmate 067)
Renalcell carcinoma (checkmate 214)
Colorectalcancer
Pancreatic?
Intravenous CD152 cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and blocks interaction with its ligands CD80/CD86
Iratumumab
MDX060
Human monoclonal antibody IgG1κ Antineoplastic Hodgkin's lymphoma phase II completed
Clinical research discontinued 2009
Intravenous CD30 (tumor necrosis factor receptor superfamily,
Member 8; TNFRSF8) aka Ki-1 Ag
Isatuximab
SAR650984
FDA review possible 2019 Chimeric monoclonal antibody IgG1κ Antineoplastic multiple myeloma
Phase I 2019
Phase II 2022
Phase III 2025
Intravenous CD 38
Iscalimab
CFZ533
Human monoclonal antibody IgG1κ Disease modifying
Potential treat autoimmune disease
Lupus nephritis phase II 2020
Myasthenia gravis
GVHD
Kidney transplant 2022 phase II
Preclinicals
Intravenous CD40
Istiratumab
MM-005
MM-141
Human monoclonal antibody IgG1 Antineoplastic
Advanced solid tumors
Pancreatic cancer phase II 2018
Insulin-like growth factor I receptor/neuregulin receptor HER3 (IGF1R, CD221)
Itolizumab Alzumab
FDA
Humanized monoclonal antibody IgG1κ Disease modifying
Psoriasis
GVHS phase II 2022
CD6
Ixekizumab Taltz Humanized monoclonal antibody Disease modifying
Phase III radiographic axial spondyloarthritis
Psoriatic arthritis
Subcutaneous IL 17A
Keliximab
Became clenoliximab IgG4
Chimeric monoclonal antibody IgG1λ Disease modifying
Chronic asthma
Rheumatoid arthritis
CD4
Labetuzumab
hMN14
CEA-Cide Humanized monoclonal antibody IgG1 Antineoplastic
Colorectal cancer
Gastrointestinal phase II 2021imagin
Phase II completed therapy 2003
Intravenous CEA
Lacnotuzumab
MCS110
Humanized monoclonal antibody IgG1κ Antineoplastic
Breast, pigmented villonodular synovitis (PVNS)
Squam ESOP phase II 2024
Gastric 2019
CSF1, MCSF
Ladiratuzumab vedotin Humanized monoclonal antibody IgG1κ
Antibody drug conjugate
Antineoplastic
Phase I triple-negative breast cancer 2017 ongoing study
LIV-1
Lampalizumab
RG7417
Humanized monoclonal fragment IgG1κ Disease modifying
Geographic atrophy secondary to age-related macular degeneration phase III Jan 2018 ineffective
Intravitreous Complement factor D (CFD)
Lanadelumab
SHP643
DX-2930
Takhzyro
FDA 2018
Human monoclonal antibody IgG1κ Disease modifying
Angioedema phase III 2019
Subcutaneous Kallikrein (KLKB1)
Landogrozumab
LY2495655
Humanized monoclonal antibody IgG4κ Disease modifying
Muscle wasting disorders, i.e., after hip surgery phase II
Pancreatic cancer phase II no benefit cancer, some muscle improvement but primary objective not met
Intravenous
Subcutaneous
Human growth differentiating factor 8 (GDF-8) aka myostatin (MSTN)
Laprituximab emtansine
IMGN289
Chimeric monoclonal antibody No trials or PubMed or creative lab only in FDA registry ?new EGFR
Larcaviximab ZMAPP Chimeric monoclonal antibody IgG1κ Disease modifying
Ebola virus
No studies clinical trial or PubMed Ebolavirus glycoprotein
Lebrikizumab
MILR1444A
TNX-650
RG-3637
PRO301444
Humanized monoclonal antibody IgG4κ Disease modifying
Asthma phase III
Atopic dermatitis HL phase II 2007
Subcutaneous injection Interleukin-13 (IL-13)
Lemalesomab Murine monoclonal antibody IgG1κ Diagnostic agent NCA-90 (granulocyte antigen)
Lendalizumab
Olendalizumab
ALXN-1007
Humanized monoclonal antibody IgGκ Disease modifying
Antiphospholipid syndrome
GI GVHD
Intravenous Anticomplement 5A
Lenvervimab Humanized IgG1κ Disease modifying
Hepatitis B
No studies in clinical trials or PubMed Hepatitis B surface antigen
Lenzilumab
KB-003
Human monoclonal antibody Antineoplastic chronic myelomonocytic leukemia and juvenile myelomonocytic leukemia phase I Intravenous GRANULOCYTEMACROPHAGE COLONY-STIMULATING FACTOR (GM-CSF)
Lerdelimumab
CAT-152
Trabio Human monoclonal antibody IgG4 Disease modifying
Phase I studies
?Cancer and fibrosis
Trials stopped for fibrosis after glaucoma surgery
Transforming growth factor β 2
Leronlimab
PRO-140
FDA review 2018 Humanized IgG4κ Disease modifying
HIV phase III ongoing no results published good results phase II
Subcutaneous Chemokine receptor 5 (CCR5)
Lesofavumab
RG70026
Human monoclonal antibody IgG1κ Disease modifying
Influenza A
No studies clinical trials or PubMed Hemagglutinin HA
Letolizumab Humanized synthetic light chain variable region (scFv) Disease modifying inflammatory diseases No studies clinical trials or PubMed or creative labs TRAP
Lexatumumab
HGS1018
HGS-ETR2
Human monoclonal antibody IgG1λ Antineoplastic
Breast
Pancreatic
Intravenous Tumor necrosis factor receptor superfamily member 10B/death receptor 5 (TRAIL-R2)
Libivirumab Humanized monoclonal antibody IgG1κ Antiinfectious
Prevent disease Hep B
Oral therapy
Abstract of randomized study of 50 patients
Hepatitis B surface antigen
Lifastuzumab vedotin
DBNIB0600A
Humanized monoclonal antibody Antineoplastic
Ovarian cancer
Phase 2
Intravenous Phosphate-sodium cotransporter
Ligelizumab
QGE031
Humanized monoclonal antibody IgG1κ Disease modifying
SSevere asthma and chronic spontaneous urticaria phase II and III ongoing trial 2021
Subcutaneous Immunoglobulin E (IGHE)
Lilotomab satetraxetan Betalutin Murine monoclonal antibody IgG1 Antineoplastic
NHL 2020/phase II 2025
Diffuse lg B-cell lymphoma 2019
CD37
Lintuzumab
SGN33
Humanized monoclonal antibody IgG1κ Antineoplastic
AMLphase I/II 2022
Mult myeloma phase I 2020
Myelodysplastic phae II 2011
Intravenous CD33
Lirilumab
IPH2102
Human monoclonal antibody IgG4 Antineoplastic
Solid and hematological cancers
No good aml, squam cell head neck no good, bladder cancer ongoing?
May benefit MDS
Intravenous Killer cell immunoglobulin like (KIR2D)
Block the interaction between KIR2DL-1,-2,-3 inhibitory receptors and their ligands
Lodelcizumab
LFU720
Humanized monoclonal antibody IgG1κ Disease modifying
Hypercholesterolemia
Unknown studies in clinical trials and PubMed Proprotein convertase subtilisin/kexin type 9 (PCSK9)
Lokivetmab Cytopoint
FDA approved for dogs only
Canis monoclonal antibody IgG2κ Disease modifying
Veterinary
Clinical signs of atopic dermatitis in dogs
Canis lupus familiaris IL31
Loncastuximab tesirine
ADCT-402
Chimeric monoclonal antibody IgG1κ Antineoplastic
Diffuse large B-cell lymphoma phase II 2020
Intravenous CD19
Lorvotuzumab mertansine
BB-10901
IMGN901
Humanized monoclonal antibody IgG1κ Antineoplastic
SCLC
Ovarian
AML phase II
Wilm, rhabdomyosarcoma, Neuroblast, MPNST, Synovial sarcoma 2018 phase II
Intravenous CD56
Losatuxizumab vedotin
ABBV-221
Chimeric/humanized monoclonal antibody IgG1 Antineoplastic Epidermal growth factor (EGRF, ERBB1 HER1)
Lucatumumab
HCD122
Discontinued development by Novartis 2013 Human monoclonal antibody IgG1κ Antineoplastic
Multiple myeloma, non-Hodgkin's lymphoma, Hodgkin's lymphoma
Intravenous CD40
Lulizumab pegol Humanized monoclonal antibody Disease modifying
SLE
Phase I safe
Phase II no response
Intravenous
Subcutaneous
CD28
Lumretuzumab
RG7116
RO5479599
Humanized monoclonal antibody IgG1κ Antineoplastic Intravenous CD28; receptor for tyrosine-protein kinase(erbB-3, HER3)
Lupartumab amadotin
BAY-1129980
Human monoclonal antibody IgG Antineoplastic
Phase I terminated Why?
Intravenous GPI- anchored cell surface-associated protein C4.4A (LYPD3)
Lutikizumab
ABT981
Humanized monoclonal antibody Disease modifying
Osteoarthritis
Phase IIa no effect
Subcutaneous Interleukin 1 alpha/interleukin 1 beta
Mapatumumab
HGS1012
Human monoclonal antibody IgG4λ Antineoplastic
Hepatocellular no benefit
Multiple myeloma
Cervical cancer
NSCLC no benefit
NHL
Bladder cancer may be beneficial
Tumor necrosis factor receptor superfamily member 10A; cytokine receptor DR4 (death receptor 4 tumor necrosis receptor apoptosis-induced ligand (TRAIL-R1)
Margetuximab
MGAH22
Chimeric/Humanized monoclonal antibody IgG1κ Antineoplastic
Breast cancer
Gastric cancer/GEC phase Ib/II trial
Intravenous erbB2/HER2
Marstacimab
PF-06741086
Human monoclonal antibody IgG1λ Disease modifying
Bleeding with hemophilia phase II 2020
Subcutaneous Tissue pathway factor inhibitor (TFPI)
Maslimomab Murine monoclonal antibody Immunosuppressive
Unknown no studies and not listed in creative lab or FDA
T-cell receptor
Matuzumab
EMD 72000
Humanized monoclonal antibody IgG1κ Antineoplastic
Colorectal, lung and stomach cancer weakly beneficial
Intravenous Epidermal growth factor receptor (EGFR)
Mavrilimumab
CAM3001
Human monoclonal antibody IgG4λ Disease modifying rheumatoid arthritis phase IIb good Subcutaneous GMCSF receptor α-chain
MEDI565
MT111
AMG211
Fab IgG1 BiTE Antineoplastic
Gastrointestinal adenocarcinoma phase I 2018
Intravenous CD3 and CEA
Mepolizumab
SB-240563
Bosatria
Nucala
FDA 2015
EU 2015
Human monoclonal IgG1κ Disease modifying
No benefit in eosinophilic esophagitis
Beneficial allergic severe asthma
Subcutaneous Interleukin-5 (IL-5) antagonist
Metelimumab
CAT 192
Humanized monoclonal antibody IgG4 Disease modifying
Scleroderma
Dropped from further development TGF β 1
Milatuzumab
HLL1
IMMU-115
Humanized monoclonal antibody IgG1κ Antineoplastic
Multiple myeloma
Lupus
Leukemia
Intravenous CD74
Minretumomab
MOAB CC49
Murine monoclonal antibody IgG1 Diagnostic
Tumor detection/diagnostic/prognostic
Failed phase I clinical trials
Tumor-associated glycoprotein 72 (TAG-72)
Mirikizumab
LY3074828
Humanized monoclonal antibody Disease modifying
Psoriasis phase III 2020
UC phase III 2023
LUCENT 1 2021 phase III
LUCENT 2 2022
Intravenous IL23A
Mirvetuximab soravtansine
M9346A
IMGN853
Chimeric monoclonal antibody IgG1 Antineoplastic
Ovarian phase III 2019
Breast ca phase II 2020
Intravenous Folate receptor alpha
Mitumomab
BEC-2
Murine monoclonal antibody Antineoplastic
SCLC phase III no benefit 2005
GD3 ganglioside
Modotuximab
1024 DS
Zatuximab
Futuximab
SYM004
Chimeric monoclonal antibody IgG1κ Antineoplastic
Antineoplastic
Colorectal
Phase 2019
Phase III 2025
Subcutaneous EGFR extracellular domain III/HER1
Mogamulizumab
AMG761
KM8761
Poteligeo
FDA 2018
Humanized monoclonal antibody IgG1κ Antineoplastic
Adult T-cell leukemia/lymphoma
Solid tumors
Many studies ongoing
Intravenous CC chemokine receptor CCR4
MOR202
MOR03087
Human monoclonal antibody IgG1 Antineoplastic multiple myeloma phase I 2018 Intravenous CD38
Monalizumab
NN8765
IPH2201
Humanized monoclonal antibody IgG4κ Disease modifying
Rheumatoid arthritis, antineoplastic gynecologic malignancies, and other cancers phase II 2021
NSCLC phase II 2022
s/p stem cell transplant phase I 2020
CLL phase II 2019
Intravenous Killer cell lectin-like receptor subfamily C member1 (NKG2A, CD159A, CD94) that recognize nonclassical HLA (i.e., HLA-E)
Morolimumab Human monoclonal antibody IgG1 ?Diagnostic No studies in pub med, creative lab or FDA substance Rhesus factor
Mosunetuzumab
RG7828
BTCT4465A
Humanized monoclonal antibody IgG1κ bispecific Antineoplastic
NHL phase II 2023
DLBCL phase II 2023
Intravenous
Subcutaneous
CD3E, MS4A1, CD20
Motavizumab
MEDI-524
Numax
FDA not approved 2010
Older drug just as effective with less side effects
Humanized monoclonal antibody IgG1κ Disease modifying
Respiratory syncytial virus phase III completed
Safety concerns hives and allergic reactions
Intramuscular Respiratory syncytial virus glycoprotein F
Moxetumomab pasudotox Lumoxiti
FDA 2018
Recombinant immunotoxin comprised of a variable fragment (Fv) of a Murine IgG4 anti-CD22 monoclonal antibody genetically
Fused to a truncated fragment of Pseudomonas exotoxin A
Antineoplastic
Hairy cell leukemia
Phase I ALL peds
Intravenous CD22
Muromonab-CD3
Muromab
Aka teplizimab/MGA031
Orthoclone OKT3
FDA 1986
EU 1986 (country specific approval)
Humanized monoclonal antibody IgG2aκ Disease modifying
Prevention of kidney transplant rejection
Many trials GVHD, NASH and T2DM, giant cell myocarditis AbATE
Intravenous
Oral
CD3
Nacolomab tafenatox Murine monoclonal fragment Fab Antineoplastic
?Colorectal cancer
No studies in clinical trial or PubMed C242 antigen
Namilumab
MT203
Human monoclonal antibody IgG1κ Disease modifying
Ank spond psoriasis, RA phase II
Subcutaneous Colony-stimulating factor 2 (CSF2)
Naptumomab estafenatox
TTS-CD3
ANYARA
ABR-217620
Murine monoclanl antibody fragment Fab Antineoplastic
Nonsmall cell lung carcinoma, renal cell carcinoma phase III completed primary endpoint not achieved
Intravenous Tumor-associated antigen 5T4
Naratuximab emtansine
IMGN529
Chimeric monoclonal antibody IgG1κ Antineoplastic
B-Cell lymphoma
NHL
Intravenous Tetraspanin-26 (CD37)
Narnatumab
IMC-RON-8
Ron8
Human monoclonal antibody IgG1κ Antineoplastic
Solid tumors phase I
Intravenous Human cell surface receptor RON (CD 135) macrophage-stimulating 1 receptor
Natalizumab
Antegran
Antegren
Tysabri
FDA 2004
EU 2006
Humanized monoclonal antibody IgG4κ Disease modifying
Relapsingmultiple sclerosis, Crohn's disease
Intravenous L selectin (CD62L)
α4-subunit of α4β1 and α4β7 integrins of leukocytes (except neutrophils) (VLA-4)
Navicixizumab
OMP 305B83
Humanized/chimeric monoclonal antibody IgG2κ Antineoplastic
Phase I study colorectal
gyn tumors
Intravenous Delta-like 4 (DLL4)
Vascular endothelial growth factor A (VEGF-A)
Navivumab
CT-P27
Human monoclonal antibody IgG1κ Disease modifying
Influenza A
No studies PubMed Influenza A virus hemagglutinin HA
Naxitamab
HU3F8
Humanized monoclonal antibody IgG3 Antineoplastic
High-risk neuroblastoma and refractory osteomedullary disease study 2023
?Intravenous c-Met
Ganglioside anti-GD2
Nebacumab Humanized monoclonal antibody IgM Withdrawn for safety,
Efficacy and commercial reasons
Endotoxin
Necitumumab
IMC-11F8
Portrazza
FDA 2015
EU 2016
Human monoclonal antibody IgG1κ Antineoplastic
Nonsmall cell lung carcinoma
Intravenous EGFR
Nemolizumab
CIM331
CD14152
Humanized monoclonal antibody IgG2κ Disease modifying
Eczema phase I and II
Subcutaneous Interleukin-31 receptor A (IL31RA)
NEOD001
Birtamimab
ELT1-01
HU2A4
Humanized monoclonal antibody IgG1κ Disease modifying
Primary systemic amyloidosis lack clinical benefit
Intravenous Amyloid A protein/amyloid light chain
Nesvacumab
REGN910
SAR307746
Human monoclonal antibody IgG1κ Antineoplastic
Solid tumors not as beneficial as other agents in breast cancer
Disease modifying
Macular degeneration
Intravenous Angiopoietin 2
Netakimab Chimeric monoclonal antibody Disease modifying
Psoriasis
PLANETA study (Russia, future EU and China)
Interleukin 17A
Nimotuzumab Theracim
Theraloc
Humanized monoclonal antibodyIgG1κ Antineoplastic
Squamous cell carcinoma, head and neck cancer, nasopharyngeal cancer, glioma
Intravenous EGFR
Nirsevimab
MEDI8897
Human monoclonal antibody IgG1κ Disease modifying
Respiratory syncytial virus phase II 2018
Intramuscular Respiratory syncytial virus fusion protein (RSVFR)
Nivolumab Opdivo
FDA 2015
EU 2015
Human monoclonal antibody IgG4κ immunoglobulin Antineoplastic agent
Programmed death receptor-1 (PD-1) blocking antibody
NSCLC, bladder cancer, renal cell cancer phase III 2021
Hodgkinlymphoma
Melanoma
Smallcell lung cancer
Squamouscarcinoma head and neck
Colorectalcancer
GBM no added benefit 2017
Intravenous Blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2
Nofetumomab merpentan Verluma
FDA 1996
No longer marketed in USA
Murine monoclonal fragment IgG2bκ Fab Cancer diagnostic imaging
SCLC
Antitumor
Membrane-spanning 4-domains, subfamily A, member 1
Obiltoxaximab
ETI-204
Anthim
FDA 2016
Chimeric monoclonal antibody IgG1κ Disease modifying
Bacillus anthracis anthrax phase IV 2021
Intravenous
Intramuscular
Bacillus anthracis spores
PA component of B. anthracis toxin
Obinutuzumab
GA101HUMAB
RG7159
RO5072759
Afutuzumab
Gazyvaro
FDA 2013
Humanized monoclonal antibody IgG1κ Antineoplastic lymphoma phase II (MCL, DLBCL)
Chroniclymphocytic leukemia
Phase II 2021
Intravenous CD20
Induces B-cell apoptosis
Ocaratuzumab
LY2469298
AME-133V
Humanized monoclonal antibody IgG1κ Antineoplastic
NHL
Pemphigus phase III
Intravenous CD20
Ocrelizumab Ocrevus
FDA 2017
Humanized monoclonal antibody IgG1κ Disease modifying
Multiple sclerosis
Intravenous CD20
Odulimomab Murine monoclonal antibody Disease modifying
Transplant rejection
Only studied in mice
Lymphocyte function-associated antigen-1 (LFA-1 (CD11a))
Ofatumumab Arzerra
FDA 2009
EU 2010
Human monoclonal antibody IgG1κ
Complement-dependent cytotoxicity (CDC)
Antineoplastic CLL
Phase III 10% ORR after ritux
Phase II as first line 86% ORR
With CHOP 100% ORR with 62% CR
Intravenous CD20
Olaratumab
IMC3G3
Lartruvo
FDA 2016
EU 2016
Human monoclonal antibody IgG1κ Antineoplastic
Sarcoma phase II 2023
Ovarian not beneficial
Intravenous Platelet derived growth factor receptor alpha (PDGF-R α)
Oleclumab
MEDI9447
Human monoclonal antibody IgG1λ Antineoplastic pancreatic phase II 2021
Colorectal cancer
Bladder cancer phase I 2020
Breast cancer phase II 2022
NSCLC phase II 2022
Intravenous? 5′-nucleotidase
CD73
Olokizumab Humanized monoclonal antibody IgG4κ Disease modifying rheumatoid arthritis
Phase I 2014 phase IIb mod results
IL6
Omalizumab
IGE25
RG3648
Xolair Humanized monoclonal antibody IgG1κ Disease modifying allergic asthma
Urticaria
Subcutaneous IgE Fc region
Omburtamab Murine monoclonal antibody IgG1κ Antineoplastic
Neuroblastoma
Phase III 2022
Intracerebroventricular treatment CD276
OMS721 Human monoclonal antibody Disease modifying
Atypical hemolytic uremic syndrome phase III 2020
Lupus nephritis phase II 2018
Intravenous Mannan-binding lectin-associated serine protease-2 (MASP-2)
Onartuzumab
PRO143966
RO5490258
METMAB
Humanized monoclonal antibody IgG Antineoplastic Intravenous Human scatter factor receptor kinase
Ontuxizumab
MORAB-004
Chimeric/humanized monoclonal antibody Antineoplastic
No clinical response
Intravenous Endosialin tumor endothelial marker-1 (TEM1)
Onvatilimab Human monoclonal antibody IgG1κ Nothing in PubMed Vista (V-domain immunoglobulin suppression of T activation (VSIR)
Opicinumab
BIIB033
Human monoclonal antibody IgG1 Disease modifying multiple sclerosis
Phase II 2020
Leucine-rich repeat and immunoglobulin domain containing neurite outgrowth inhibitor receptor interacting protein-1 (LINGO-1) LINGO-1
Oportuzumab monatox
VB4-845
Vicinium
Proxinium
FDA 2005
EU 2005
Additional approval pending 2019
Humanized monoclonal antibody fragment
scFv
Antineoplastic
Bladder phase III
Headand neck cancer
Intravescical Epithelial cell adhesion molecule (EPCAM) and tumor-associated calcium signal transducer 1 (TACSTD1) and pseudomonas exotoxin A immunotoxin fusion protein (anti-EPCAM antibody fragment-Pseudomonas exotoxin fusion protein)
Oregovomab
MAB-B43.13
OvaRex Murine monoclonal antibody IgG1κ
Antiidiopathic antibody to ovarian antigen CA-125
Antineoplastic
Ovarian cancer
Not effective in achieving increase RFS or OS
Ovarian phase I 2021
Phase II 2019
Subcutaneous
Intravenous
CA-125
Orticumab
RG7418
Human monoclonal antibody fragment Fab Disease modifying
Antiinflammatory
Oxidized low-density lipoprotein oxLDL
Otelixizumab Chimeric humanized monoclonal antibody IgG1 Disease modifying
Diabetes mellitus type 1
TTEDD phase II
DEFEND-1 phase III failed
DEFEND-2 phase III- no real benefit
Subcutaneous CD3
Otilimab
MOR103
GSK3196165
Human monoclonal antibody IgG1λ Disease modifying
Osteoarthritis, rheumatoid arthritis phase II 2012
Multiple sclerosis phase II 2014
Intravenous Granulocyte-macrophage colony-stimulating factor (GMCSF)
Otlertuzumab
TRU-016
Humanized monoclonal antibody IgG fragment Antineoplastic
CLL phase I and II 2014 and 2019
Intravenous CD37
Oxelumab
OX40L
R4930
HUMAB OX40L
Human monoclonal antibody IgG1κ Disease modifying
Asthma mainly preclinical mice
Many clinical studies ongoing leukemia and asthma
Intravenous OX-40 (CD252)
Ozanezumab
GSK1223249
Humanized IgG1 Disease modifying
ALS phase II 2015 ALS no good
Intravenous Neurite outgrowth inhibitor (NOGO-A)
Ozoralizumab
ATN 103
Humanized monoclonal antibody Disease modifying
Rheumatoid arthritis phase II 2012
Subcutaneous TNF-α
Pagibaximab Chimeric monoclonal antibody Disease modifying
Staph sepsis low birth weight infants
Phase II/III studies 2010
Intravenous Lipoteichoic acid
Palivizumab Synagis, Abbosynagis
FDA 1998
EU 1999
Humanized monoclonal antibody IgG1κ Disease modifying
RSV many phase III studies
Intramuscular F protein of respiratory syncytial virus
Pamrevlumab
FG-3019
Human monoclonal antibody IgG1κ Disease modifying
Idiopathic pulmonary fibrosis (IPF),
Antineoplastic
Pancreatic cancer
Muscular dystrophy phase II 2021
Diabetes nephropathy
Connective tissue growth factor (CTGF)
Insulin-like growth factor binding protein 8 (IGFBP-8)
Panitumumab
ABENIX
ABX-EGFhttps://en.wikipedia.org/wiki/List_of_therapeutic_monoclonal_antibodies - cite_note-WHOList91-38
Vectibix
FDA 2006
EU 2007
Human monoclonal antibody IgG2κ Antineoplastic
Metastaticcolorectal cancer
Intravenous EGFR/erbB-1/HER1
PankoMab-GEX
Gatipotuzumab
Humanized monoclonal antibody IgG1κ Antineoplastic
Phase IIb 2017
Phase I solid tumors 2019
Intravenous Tumor-specific glycosylation of MUC1
Panobacumab
Aerumab 11
AR-101
KBPA-101
Human monoclonal antibody Antimicrobial
Pseudomonas aeruginosa infection
Intravenous Pseudomonas aeruginosa serotype O11
Parsatuzumab
MEGF0444A
RG-7414
Human monoclonal antibody IgG1κ Antineoplastic
Colorectal cancer phase II 2014 no benefit
Epidermal growth factor-like domain 7 (EGFL7)
Pascolizumab Humanized monoclonal antibody Disease modifying
Not effective trails aborted
IL-4
Pasotuxizumab Chimeric/humanized monoclonal antibody fragment Antineoplastic No studies Folate hydrolase/prostate-specific membrane antigen (PSMA)
Pateclizumab
RG7415
PRO283698
MLTA3698A
Humanized monoclonal antibody IgG1κ Disease modifying rheumatoid arthritis
Phase II not as efficacious as adalimumab but had response
Subcutaneous Lymphotoxin-α
Patritumab
AMG888
U3-1287
Human monoclonal antibody IgG1κ Antineoplastic
May not be beneficial head/neck NSCLC CT site
ErbB3 (HER3)
Spartalizumab
PDR001
FDA review possible 2019 Humanized monoclonal antibody Antineoplastic
Breast cancer phase II 2021
NSCLC 2021
Melanoma phase II 2022
Phase III 2020
Intravenous PD1, PDCD1, CD279
Pembrolizumab
MK-3475
Keytruda
FDA 2014
EU 2015
FDA 2018 for metastatic Merkel cell carcinoma, HCC, NSCLC
Humanized monoclonal antibody IgG4κ Antineoplastic
Squamous carcinoma trachea, NSCLC, urothelial (HCC phase II)
Melanoma
cHL, LgB cell lymph
Gastric cancer
Cervicalcancer
Hepatocellular carcinoma
Intravenous
Trials for multiple myeloma discontinued by FDA
PD-1
Pemtumomab
HMFG1 antibody labeled with 90Yttrium
Theragyn Murine monoclonal antibody Antineoplastic
Phase III Europe 2009/US 2013 no benefit after 3.5 years follow-up
MUC1/human milk fat globule antigen 1 (HMFG1)
Perakizumab Humanized monoclonal antibody IgG1κ Disease modifying psoriatic arthritis
Phase I discontinued
IL 17A
Pertuzumab Perjeta
FDA2012
EU 2013
Omnitarg
Humanized monoclonal antibody IgG1 Antineoplastic agent
HER2-positive metastatic breast cancer
Gastric/breast cancer
Phase III gastric
Intravenous Extracellular dimerization domain (subdomain II) of the human epidermal growth factor receptor 2 protein (HER2/neu)
Pexelizumab Humanized scFv Disease modifying acute myocardial infarctions APEX-AMI trial negative results
PRIMO-CABG I and II trials no significant benefit
C5
Pidilizumab
CT-011
Humanized monoclonal antibody IgG1κ Antineoplastic
Mult myeloma
DLBCL
Pontine glioma
Pancreas
Melenaoma
HCC
Antiinfection
Intravenous PD-1
Pinatuzumab vedotin Humanized monoclonal antibody
ADC consisting of the microtubule-disrupting agent, monomethyl auristatin E (MMAE), conjugated to an anti-CD22 mAbvia the protease-cleavable peptide linker maleimidocaproylvaline-citrulline(vc)-p-aminobenzoyloxycarbonyl
Antineoplastic B-cell NHL phase I study good response
Phase II completion 2019
Intravenous CD22
Pintumomab Murine monoclonal antibody Not therapeutic
Diagnostic imaging adenocarcinoma antigen
Adenocarcinoma (imaging)
Placulumab Human monoclonal antibody V-kappa)2 FC Disease modifying pain and inflammatory diseases
Development discontinued 2012
Human TNF
Plozalizumab
MLN1202
HU1D9
Withdrawn by company Humanized monoclonal antibody IgG1κ Disease modifying
Diabetic nephropathy and arteriovenous graft patency
RA no benefit
Intravenous CC chemokine receptor 2 (CCR2)
Pogalizumab
MOXR0916
R07021608
Vonlerolizumab
Humanized monoclonal antibody IgG1κ Antineoplastic
Solid tumors phase I 2019 may be safe but may not be effective
No formal manuscripts yet
Intravenous Tumor necrosis factor receptor superfamily member 4 (ACT35, OX40, CD134)
Polatuzumab vedotin
FCU2711
RO5541077-000
FDA review 2018 Humanized monoclonal antibody IgG1κ Antineoplastic
NHL phase II 2019
DLBCL phase III 2023
Intravenous CD79B
Ponezumab
RN1219
PF-04360365
Humanized monoclonal antibody IgG2 Disease modifying Alzheimer's disease
Safe but no clinical efficacy 2013
Intravenous Human beta-40-amyloid Aβ40
Porgaviximab
C2G4
Chimeric monoclonal IgG1κ Antiinfectious
Ebola virus disease
No known ongoing studies Zaire ebolavirus glycoprotein
Prasinezumab
PRX002
RG7935
RO7046015
Humanized monoclonal antibody IgG1κ Disease modifying
Parkinson's disease
Phase II 2021
Intravenous Anti-alpha-synuclein (NACP)
Prezalizumab
AMG-557
MEDI5872
Humanized monoclonal antibody IgG2 Disease modifying
SLE phase II 2018
Sjogren's
Subcutaneous B7-related protein inducible T-cell costimulator ligand (ICOSL)
Priliximab
cMT 412
CEN 000029
Chimeric monoclonal antibody Disease modifying
Crohn's disease, multiple sclerosis
IN FDA no known studies CD4
Pritoxaximab Chimeric monoclonal antibody IgG1κ Antiinfectious E. coli shiga toxin type-1
Pritumumab Human monoclonal antibody IgG1κ Antineoplastic
Brain cancer
Phase II studies in Japan, could not find literature reportedly increase survivability 10 fold
Vimentin
Quilizumab
MEMP1972A
RG-7449
Anti-M1
Humanized monoclonal antibody IgG1κ Disease modifying
Asthma phase II 2014 no great benefit
Urticaria phase II 2014 no great benefit
Allergic rhinitis
Subcutaneous
Intravenous
M1 prime segment of membrane bound IgE (IGHE)
Racotumomab Vaxira Murine monoclonal antibody IgG1κ Antineoplastic
Nonsmall cell lung cancer phase III 2016 cimavax better (recombinant EGF injection) 2 more months survival over placebo
Neuroblastoma phase II 2020
Intradermal
Subcutaneous
N-glycolylneuraminic acid gangliosides (NGNA ganglioside)
Radretumab
F16SIP
L19SIP radiolabeled with I331
Human monoclonal antibody
Imaging study PET
Antineoplastic
Lymphoma brain mets 2012 phae I
Stage III NSclC
Fibronectin extra domain-B
Rafivirumab
CR57
Human monoclonal antibody IgG1λ
Used in cocktail and with vaccination
Antiinfectious
Rabies (prophylaxis)
No known studies Rabies virus glycoprotein
Ralpancizumab
RN317
PF-05335810
Humanized monoclonal antibody IgG2κ Disease modifying
Dyslipidemia phase I 2017
PCSK9 (proprotein convertase subtilisin/kexin type 9, neural apoptosis-regulated convertase 1, NARC1, NARC-1, proprotein convertase 9, PC9)
Ramucirumab
LY3009806
IMC-1121B
Cyramza
FDA 2014
EU 2014
Human monoclonal antibody IgG1κ Antineoplastic
Urothelial phase III done
Adenocarcinomastomach and GE junction phase II 2023
Colorectal cancer
NSCLC
HCC phase III 2017 no additional benefit
Intravenous VEGFR2
Ranevetmab
NV-01
Veterinary monoclonal antibody IgG1κ canine Disease modifying
Osteoarthritis in dogs
Nerve growth factor-β (NGF-β)
Ranibizumab
RBZ
RG-3645
RHuFAb
Lucentis
FDA 2006
EU 2007
Humanized monoclonal fragment IgG1κ Fab Disease modifying
Maculardegeneration (wet form) post market studies phase II
Intravitreal Vascular endothelial growth factor A (VEGF-A)
Ravagalimab
PR-1629977
ABBV-323
Humanized monoclonal antibody IgG1κ Disease modifying UC phase II 2023 Intravenous
Subcutaneous
CD40
Ravulizumab
ALXN1210
Ultomiris
FDA 2019
EU pending
Humanized monoclonal antibody IgG2/IgG4κ Disease modifying
Paroxysmal nocturnal hemoglobinuria (PNH)
Phase III 2021 similar to eculizumab, atypical hemolytic uremic syndrome phase III 2021
Intravenous Complement C5 (C5)
Raxibacumab ABthrax
FDA 2012
Human monoclonal antibody IgG1λ Antiinfectious
Treat inhalation anthrax
Intravenous Bacillus anthracis protective antigen
Refanezumab
GSK249320
Humanized monoclonal antibody IgG1κ Disease modifying recovery of motor function after stroke
Phase II completed 2011 no benefit
Intravenous Myelin-associated glycoprotein
Regavirumab
MCA C23
TI-23
Human monoclonal antibody Antiinfectious
Cytomegalovirus glycoprotein B
ONLY STUDIES IN RATS 1994
Cytomegalovirus infection
Relatlimab
BMS-986016
Human monoclonal antibody IgG4κ Antineoplastic
Melanoma phase II 2022
Colon cancer phase II 2022
Chordoma phase II 2020
Cannot find manuscripts but company website phase II good results
Glioblastoma phase I 2020
Intravenous Lymphocyte activation gene 3 (LAG3) CD223
Remtolumab
ABT-122
Human monoclonal antibody Disease modifying
RA
Phase II 2016 no increased benefit over adalimumab
Subcutaneous Interleukin 17 alpha, TNF-α
Reslizumab
DCP 835
Scheme 55700
CEP-38072
Cinqair
FDA 2016
EU 2016
Humanized monoclonal antibody IgG4κ Disease modifying
Inflammations of the airways asthma completed and ongoing, skin and gastrointestinal tract, polyarteritis stage II 2018
Rhino sinusitis 2020
Intravenous
Subcutaneous
IL-5
Rilotumumab
AMG-102
Human monoclonal antibody IgG2κ Antineoplastic
Gastric completed phase III 2015 not effective
NSCLC phase II 2014 no benefit
Glioma phase II no response
Intravenous Hepatocyte growth factor (HGF)
Rinucumab
REGN2176
Human monoclonal antibody IgG4κ Disease modifying neovascular age-related macular degeneration phase II 2014 Intravitreal Platelet-derived growth factor receptor beta
Risankizumab
ABBV-066
BI-655066
FDA/EU pending approval Humanized monoclonal antibody IgG1κ Disease modifying Crohn's disease phase II good, phase III ongoing
psoriasis phase II response better than ustekinumab, psoriatic arthritis, and asthma
Subcutaneous IL23A
Rituximab
GP2013
IDEC-102
RG-105
MabThera, Rituxan
FDA 1997
EU 1998
Chimeric monoclonal antibody IgG1κ Antineoplastic
Non-Hodgkin lymphomas, chronic lymphocytic leukemias, some autoimmune disorders, i.e., rheumatoid arthritis, >2K studies ongoing
Subcutaneous CD20
Rivabazumab pegol Humanized monoclonal antibody fragment Fab’ IgG1κ Antiinfectious No studies found Pseudomonas aeruginosa type III secretion system
Rmab Rabishield
Made in India
Human monoclonal antibody Antiinfectious
Postexposure prophylaxis of rabies
Rabies virus G glycoprotein
Robatumumab
19D12
SCH 717454
MK-7454
P04722
Human monoclonal antibody IgG1κ Antineoplastic
Colorectal phase II 2009 little benefit
Ewings no response 2016
Intravenous Insulin-like growth factor I (IGF-1 receptor) (CD221)
Roledumab Human monoclonal antibody IgG1κ Immunomodulation
Rh disease
Phase III 2017
Intravenous RHD
Romilkimab
SAR156597
HUBTI3_2_1
Humanized chimeric monoclonal antibody IgG4 bispecific Disease modifying
Systemic sclerosis phase II 2019
Pulm fibrosis phase II 2017 no benefit
Subcutaneous Interleukin 13 and IL4
Romosozumab Evenity
FDA pending
EU pending
Japan 2018
Humanized monoclonal antibody IgG2κ Disease modifying
Postmenopausal osteoporosis phase III study FRAME
Men phase III
BRIDGE phase III
Intravenous Sclerostin/scleroscin SOST
Rontalizumab
rhuMAb IFNalpha
Humanized monoclonal antibody Disease modifying
Systemic lupus erythematosus phase II 2013 end points not met
Subcutaneous IFN-α
Rosmantuzumab
OMP-131R10
Humanized monoclonal antibody IgG1κ Antineoplastic
Colorectal cancer phase I 2018
Intravenous Root plate-specific spondin r-spondin-3
WNT? (wingless/integrated)
Rovalpituzumab tesirine
SC0002
SC16LD6.5
ABBV-181
Humanized monoclonal antibody IgG1κ Antineoplastic
Small cell lung cancer phase I 2018
Phase II 2024
Intravenous Delta-like ligand-3 (DLL3)
Rovelizumab
Hu23F2G
LeukArrest Humanized monoclonal antibody IgG1κ Disease modifying
Hemorrhagic shock, MI stroke phase III goals not met 2000
CD11, CD18
Rozanolixizumab
UCB7665
Chimeric/humanized monoclonal antibody IgG4κ Thrombocytopenia ITP phase II 2019
Myasthenia gravis phase II 2018
Subcutaneous
Intravenous
Neonatal Fc receptor (FCGRT)
Ruplizumab Antova Humanized monoclonal antibody Disease modifying lupus and lupus nephritis not effective
Life-threatening thromboembolism
BioDrugs. 2004; 18(2):95–102.
Costimulation blockade in the treatment of rheumatic diseases
CD154 (CD40L)
Sacituzumab govitecan
IMMU-132
FDA/EU pending approval Humanized monoclonal antibody IgG1κ Antineoplastic agent
Prostate cancer phase II 2021
Urothelial phase II 2020
Trip neg breast ca phase III 2020 NSCLC SCLC UC
Intravenous Tumor-associated calcium signal transducer 2 (TROP-2) inhibits topoisomerase I
Samalizumab
ALXN6000
BAML-16-001-S1
Humanized monoclonal antibody IgG2/G4κ Antineoplastic
CLL MM phase I 2010 (terminated by sponsor)
AML phase II 2021
Intravenous OX-2 membrane glycoprotein (CD200)
Samrotamab vedotin Chimeric/humanized monoclonal antibody IgG1κ Antineoplastic No studies found Leucine-rich repeat-containing protein 15 (LRRC15)
Sarilumab
REGN88
SAR153191
Kevzara
FDA?EU/Japan under review approved in Canada
Human monoclonal antibody IgG1κ Disease modifying rheumatoid arthritis phase III 2015/2020/2027(preg exposure), ankylosing spondylitis
Juvenile idiopathic arthritis phase II 2022
Subcutaneous IL6
Satralizumab
SA237
Sapelizumab
FDA review possible 2019 Humanized monoclonal antibody IgG2κ Disease modifying
Neuromyelitis optica phase III 2019/2020
Subcutaneous? IL6 receptor
Satumomab pendetide OncoScint CR103
FDA 1992
Murine monoclonal antibody IgGκ fragment Fab’ Diagnostic imaging
Detection colorectal and ovarian cancer
Intravenous Tumor-associated glycoprotein (TAG-72)
Secukinumab
AIN457
Cosentyx
FDA 2015
EU 2015
Human monoclonal antibody IgG1κ Disease modifying
Uveitis, rheumatoid arthritis psoriasis phase II 2019 over 100 other studies arthritis; psoriatic psoriasis; spondylitis; ankylosing
Subcutaneous IL 17A
Selicrelumab
CP 870.893
RG7876
RO-7009789
Human monoclonal antibody IgG2κ Antineoplastic
Solid tumors phase I 2020
Pancreatic cancer phase II 2020
Colon cancer phase II 2021
Mesothelioma phase ib 2015
Subcutaneous
Intravenous
Tumor necrosis factor receptor superfamily member 5 (CD40)
Seribantumab
MM121
SAR256212
Human monoclonal antibody IgG2λ Antineoplastic
Breast phase II 2020
Ovarian phase I 2014
Intravenous Receptor tyrosine-protein kinase erbB-3 (HER3)
Setoxaximab Chimeric monoclonal antibody IgG1κ Antiinfection
E. coli
No known studies or clinical use E. coli shiga toxin type-2
Setrusumab
BPS804
MOR05813
Human monoclonal antibody IgG2 Disease modifying
Osteogenesis imperfecta phase II 2020
Intravenous Sclerostin (SOST)
Sevirumab
MSL-109
Antiinfectious
CMV retinitis early termination trial secondary to safety
Phase II 2003
Cytomegalovirus infection
SHP647
Ontamalimab
PF-00547659
Human monoclonalantibodyIgG2κ Disease modifying
Crohn's/UC phase III 2020–2025 × 7
Phase II study 2007 better response in UC than in Crohn (?more time needed to evaluate clinical significance
Subcutaneous Mucosal addressin cell adhesion molecule (MADCAM)
Sibrotuzumab
BIBH1
F19
Humanized monoclonal antibody IgG1κ Antineoplastic
Colorectal cancer phase II 2003 failed
Lung cancer2001
Intravenous FAP
Sifalimumab
MDX-1103
MEDI-545
CP145
Humanized monoclonal antibody IgG1κ Disease modifying
SLE phase II 2015 dermatomyositis, polymyositis
Intravenous
Subcutaneous
IFN-α
Siltuximab
CLLB8
CNTO-328
Sylvant
FDA 2014
EU 2014
Chimeric monoclonal antibody IgG1κ Antineoplastic
Multiple myeloma phase II 2019
DM type I phase I 2017
Schizophrenia adjunct 2020 phase II
MulticentricCastleman's disease (MCD) with HIV negative and HHV-8 negative
Intravenous IL-6
Simtuzumab
AB0024
GS-6624
Humanized monoclonal antibody IgG4κ Disease modifying
Hepatic fibrosis
Phase II 2016 no benefit
Pulm fibroses phase II 2017 no benefit
Myelo fibr 2017 phase II
Subcutaneous
Intravenous
Lysyl oxidase homolog 2 (LOXL2)
Siplizumab
MEDI-507
Humanized monoclonal antibody IgG1κ Antineoplastic CD2
T Or NK cells
Sirtratumab vedotin Human monoclonal antibody Antineoplastic Nothing in PubMed or clinical trials SLITRK6
Sirukumab Human monoclonal antibody IgG1κ Disease modifying
Rheumatoid arthritis
Phase III done good results
Subcutaneous IL-6
Sofituzumab vedotin Humanized monoclonal antibody Antineoplastic
Ovarian pancreatic
Phase I (2014)
CA-125
Solanezumab
LY2062430
Humanized monoclonal antibody IgG1 Disease modifying Alzheimer's
Phase III study discontinued no effect
In preclinical trial for secondary prevention 2022
Hereditary AD phase III 2021
Intravenous Beta amyloid
Solitomab
MT110-011
AMG110
Murine monoclonal antibody bispecific T-cell engager (BiTE) Antineoplastic
Gastrointestinal, lung, and other cancers
Phase I 2015
Intravenous Epithelial cell adhesion molecule (EpCAM) CD3
Sonepcizumab
LT1009
iSONEP Humanized monoclonal antibody Disease modifying
Choroidal and retinal neovascularization phase II 2015 not so good
Antineoplastic phase II renal cancer 2017 potential
Intravenous
Intravitreous
Sphingosine-1-phosphate (S1P)
Stamulumab Humanized monoclonal antibody Disease modifying muscular dystrophy
Animal studies, minimal efficacy
Phase I/II studies ongoing (no improvement)
Intravenous Myostatin
Sulesomab
IMMU-MN3
LeukoScan
EU 1997
Murine monoclonal IgG1 fragment Fab′ Diagnostic
Osteomyelitis (imaging)
NCA-90 (granulocyte antigen)
Suptavumab
REGN2222
SAR438584
Human monoclonal antibody IgG1κ Antiinfectious
Medically attended lower respiratory disease phase III 2017 not meet primary endpoint
Another study no data yet at 30 mg/kg dose
Intramuscular Resp sync virus fusion protein (RSVFR)
Sutimlimab
BIVV009
Chimeric/humanized monoclonal antibody IgG4κ Disease modifying cold agglutinin disease phase III 2020 Intravenous Complement C1s (C1s)
Suvizumab
KD-247
Humanized monoclonal antibody IgG1κ Antiinfectious
HIV
Phase I KD-247
2007
Intravenous Human immunodeficiency virus glycoprotein 120 third variable loop
Suvratoxumab
MEDI4893
Human monoclonal antibody IgG1κ Disease modifying
Nosocomial pneumonia phase II 2018
Intravenous Staphylococcus aureus alpha toxin
Tabalumab
LY2127399
Human monoclonal lantibodyIgG4κ Antineoplastic
Rheum arthr phase III 2013 no signif response
SLE phase III 2015 endpoints not met
Mult myelo phase I 2014 may not treat but be prognostic
Subcutaneous Cytokine B-cell activating factor (BAFF)
Tacatuzumab tetraxetan
HAFP-31
AFP-Cide Humanized monoclonal antibody yttrium77 Antineoplastic No studies in clinical trial or PubMed Alpha-fetoprotein
Tadocizumab
C4G1
YM337
Humanized monoclonal antibody fragment IgG1κ Fab’ Disease modifying
Percutaneous coronary intervention phase I 1999
?not further developed
Integrin αIIbβ3
Talacotuzumab
CSL362
JNJ-56022473
Humanized monoclonal antibody IgG1-2κ Antineoplastic
AML phase III 2018
MDS phase II 2019
SLE 2019 phase I
Intravenous Interleukin 3 receptor subunit-α (IL3Rα, CD123)
Talizumab
C21/AL-90
TNX-901
Humanized monoclonal antibody IgG1κ Disease modifying
Peanut allergy
Allergic reaction
Phase II 2003 good results legal issues shelved the drug
Subcutaneous IgE
Tamtuvetmab
AT-005
Tactress Canine monoclonal antibody IgG2λ CD52
Tanezumab
RN624
PF-4383119
FDA review possible 2019 Humanized monoclonal antibody IgG2 Disease modifying
Pain
Osteoarthritis
Back pain
Metastatic cancer pain
Phase II ∼2008
Nerve growth factor (NGF)
Tanibirumab
Olinvacimab
TTAC-0001
Human monoclonal antibody IgG1 Disease modifying
Antineoplastic
Phase I glioblastoma 2020
Breast cancer phase I 2020
AMD murine no human studies found
Intravenous VEFR-2
Taplitumomab paptox Murine monoclonal antibody IgG1κ Antineoplastic No studies pub med or clinical trials CD19
Tarextumab
OMP-59R5
Human monoclonal antibody IgG2 Antineoplastic
Phase II trial NSCLC no benefit 2017
Pancreatic phase II 2017
Intravenous Notch2/3, Notch receptor
Tavolimab
MEDI0562
Chimeric/humanized monoclonal antibody IgG1κ Antineoplastic
Head and neck phase I 2024
Ovarian cancer phase II 2023
Tumor necrosis factor receptor superfamily member 4 (TNFRS4) OX40L receptor (CD134)
Technetium (99 mTc) acritumomab Rabbit monoclonal IgG Not for use in humans- research purpose only CEA
Technicium (99 mTc) Fanolesomab NeutroSpec
FDA2004
Murine monoclonal IgM radiolabeled Disease modifying osteomyelitis
Sales and marketing suspended (2005)
Diagnosticscans for acute appendicitis
Intravenous CD15
Tefibazumab
INH–H2002
Aurexis Humanized monoclonal antibody IgG1κ Antiinfectious
Staphylococcus aureus infection
Phase II 2006
Intravenous Clumping factor A
Telimomab aritox
(TAB-885)
Recombinant murine monoclonal antibody Fab with ricin Antineoplastic
T Cell lymphoma/leukemia
No studies in pub med or clinical trials CD5
Telisotuzumab vedotin
ABT-700
Humanized monoclonal antibody IgG1κ Antineoplastic
Phase I 2017
SCLC phase II 2022
NSCLC phase II 2021
Intravenous Hepatocyte growth factor receptor HGFR
Tenatumomab Murine monoclonal antibody IgG2b Antineoplastic
Phase I 2017
Phase II brain tumors 2010
Intravenous P24821, tenascin C
Teneliximab Chimeric monoclonal antibody IgG1 Not in clinical trials 2009 CD40 (TNF receptor superfamily member 5)
Teplizumab
MGA031
PRV-031
hOKT3g1(Ala-Ala)
Humanized monoclonal antibody IgG1κ Disease modifying type I DM phase II completion AbATE trial 2019
Psoriasis phase I and II completed 2010 study stopped secondary to injection reaction severe allergy
Intravenous
Subcutaneous
CD3
Tepoditamab Human monoclonal antibody IgG1κ bispecific Antineoplastic No studies on PubMed or clinical trials c-type dendritic cell-associated lectin 2 (CLEC-2A, MCLA-117) and CD3
Teprotumumab
RV001
R-1507
RO4858696
HZN-001
FDA review possible 2019 Human monoclonal antibody Disease modifying
Thyroid eye disease phase II 2017
Graves phase III 2020
Intravenous Insulin-like growth factor receptor type I (IGF-1 receptor) (CD221)
Tesidolumab
LFG316
NOV-4
Human monoclonal antibody Phase I 2017
PNH phase II 2020
AMD phase II 2015 not beneficial
Intravenous
Intravitreous
C5
Tetulomab tetraxetan LU-177 Betalutin Humanized monoclonal antibody Antineoplastic
Animal studies 2013
CD37
Tezepelumab
MEDI9929
AMG-157
Human monoclonal antibody IgG2λ Disease modifying
Asthma, atopic dermatitis
Phase II 2017
Subcutaneous Thymic stromal lymphopoietin (TSLP)
Theralizumab
TGN1412
TAB08
Humanized monoclonal antibody Antineoplastic
Solid tumors phase I 2020
Disease modifying
Rheum arth, SLE phase II
Intravenous CD28
History of cytokine storm at higher doses 2006
Tibulizumab
LY3090106
Humanized monoclonal antibody bispecific tetravalent Disease modifying
Autoimmune disorder
Phase I 2020
Subcutaneous
Intravenous
No manuscripts found specific to this antibody
Human B-cell activating factor of the tumor necrosis factor family interleukin 17 (BAFF)
Tigatuzumab
CS-1008
TRA-8
Humanized monoclonal antibody IgG1κ Antineoplastic
Colon phase II 2011no added benefit
Colon phase I 2013
NSCLC phase II 2011 no benefit
Pancreatic phase II 2008 benefit
TN breast canc 2015 phase II no added benefit
Cytokine receptor DR5 (death receptor 5) TRAIL-R2
Tildrakizumab
MK-3222
SCH-900222
Ilumya
Ilumetri
FDA 2018
Humanized monoclonal antibody IgG1κ Immunologically mediated inflammatory disorders
Mod/severe psoriasis phase III 2018-20
Subcutaneous IL23
Timigutuzumab Humanized monoclonal antibody IgG1κ Antineoplastic No studies in clinical trial or PubMed erbB2/HER2
Timolumab
BTT-1023
Human monoclonal antibody Disease modifying
Scler cholang phase II 2019
Intravenous AOC3
Tiragotumab
MTIG-7192A
RG6058
RO7092284
Human monoclonal antibody IgG1κ Antineoplastic
Phase I 2020
NSCLC phase II 2021
HL phase II 2019
Intravenous
Nothing published yet
T-cell IG and immune-receptor tyrosine-based inhibitory motif (TIGIT)
Tislelizumab China pending approval Humanized monoclonal antibody Antineoplastic
NSCLC phase III 2020
Gastric phase III 2022
Esophageal cancer phase III 2021
NHL phase II 2020
Intravenous
Nothing published yet 2019
PCDC1, CD279
Tisotumab vedotin Human monoclonal antibody IgG1κ Antineoplastic
Ovary cancer
Cervix cancer
Endometrium cancer
Bladder cancer
Prostate cancer
Esophagus cancer
Lung cancer, NSCLC
Squamous cell carcinoma of the head and neck
Pancreatic phase II 2022/3
Intravenous Coagulation factor III
Tocilizumab
MRA
R-1569
RG-1569
RHPM-1
RO-4877533
Atlizumab
Actemra, RoActemra
FDA 2010
EU 2009
Humanized monoclonal antibody IgG1κ Disease modifying rheumatoid arthritis
>100 studies
Behcet syndrome
Intravenous
Subcutaneous
IL-6 receptor
Tomuzotuximabfibri Humanized monoclonal antibody IgG1κ Antineoplastic
Phase I 2019
EGFR, HER1
Toralizumab
E−6040
IDEC-131
Humanized monoclonal antibody IgG1κ Disease modifying rheumatoid arthritis, lupus nephritis etc.
Phase II trials failed with TE
CD154 (CD40L)
Tosatoxumab Human monoclonal antibody IgG1λ Antiinfectious No studies PubMed or Clin trials Staphylococcus aureus α-hemolysin
Tositumomab
and iodine 131 Tositumomab
Bexxar
FDA 2003
Murine monoclonal antibody IgG2aλ Antineoplastic
Follicular lymphoma (NHL)
>100 studies
Intravenous CD20
Tovetumab
MEDI-575
Human monoclonal antibody IgG2κ Antineoplastic
Phase I/II 2012
Glioblastoma limited clin activity
Intravenous Platelet-derived growth factor receptor α (CD140a)
Tralokinumab
CAT-354
Human monoclonal antibody IgG4 Disease modifying asthma phase IIb +/−, atopic dermatitis phase II 2016 Intravenous
Subcutaneous
IL-13
Trastuzumab
4D5v8
R-597
SYD977
Herceptin
FDA 1998
EU 2000
Herceptin Hylecta FDA 2019 –trastuzumab/hyaluronidase
Herzuma 2018
Humanized monoclonal antibody IgG1κ Antineoplastic
Breastcancer
Gastricand gastro-esophageal junction cancer HER2-positive phase III
Subcutaneous
Intravenous
HER2/neu
Trastuzumab Deruxtecan DS-8201 Hercion
FDA breakthrough therapy
Antibody drug conjugate humanized antibody IgG1κ with topoisomerase I inhibitor (DXd) Antineoplastic breast cancer phase I study breast, gastric, colorectal, salivary, and nonsmall cell lung cancer participated in part 2 2020
phase II DESTINY-Breast01
HER2
Trastuzumab emtansine
RG-3502
PRO132365
Kadcyla
FDA2013
EU 2013
Humanized monoclonal antibody IgG1κ as ADC Antineoplastic
Breastcancer
Intravenous HER2/neu
TRBS07 Ektomab 3funct Antineoplastic
Melanoma
GD2 ganglioside Tribbles-related protein (TRB) family members are the mammalian orthologs of Drosophila tribbles. Tribbles was originally identified as a cell cycle regulator during Drosophila development. Tribbles genes are evolutionary conserved, and three TRB genes (TRB1, TRB2 and TRB3) have been identified in mammals. TRBs are considered pseudokinases because they lack an ATP binding site or one of the conserved catalytic motifs essential for kinase activity. Instead, TRBs play important roles in various cellular processes as scaffolds or adaptors to promote the degradation of target proteins and to regulate several key signaling pathways. Recent research has focused on the role of TRBs in tumorigenesis and neoplastic progression. In this review, we focus on the physiological roles of TRB family members in tumorigenesis through the regulation of the ubiquitin-proteasome system and discuss TRBs as biomarkers or potential therapeutic targets in cancer
Tregalizumab
BT-061
Humanized monoclonal antibody IgG1κ Disease modifying
Rheumatoid arthritis
Phase IIb no benefit
Subcutaneous CD4
Tremelimumab
(aka ticilimumab)
∗CP-675,206 Human monoclonal antibody IgG2 Antineoplastic agent
NSCLC, small cell lung cancer, urethelial cancer phase II 2020, head and neck cancer and colon phase I 2021
Mesothelial phase IIb DETERMINE not beneficial
>100 studies
CTLA4 (cytotoxic T lymphocyte-associated antigen 4, CD152
Trevogrumab
REGN1033
SAR391786
Human monoclonal antibody IgG4κ Disease modifying
Muscle atrophy due to orthopedic disuse and sarcopenia phase II 2020
Myostatin, growth differentiation factor 8 (GDF8)
TRL3d3
3D3
IgG Studies only in mice to this point Ati-G protein antibody (RSVGV)
Tucotuzumab celmoleukin
EMD-273066
HUKS-IL2
Humanized monoclonal antibody IgG1 Antineoplastic
Ovarian phase II 2008
Lung, kidney, bladder phase I 2000 no benefit
Intravenous Interleukin2 (EpCAM)
Tuvirumab Humanized monoclonal antibody Antiinfectious
Not effective in achieving primary efficacy as assessed by neutralization of circulating HBsAg
Intravenous Hepatitis B virus surface antigen
Ublituximab
TG-1101
FDA review pending 2019 Chimeric monoclonal antibodyIgG1κ Antineoplastic
Chronic lymphocytic leukemia, follicular cell lymphoma phase II 2020
Disease modifying
Multiple sclerosis phase II 2019, phase III 2021
Awaiting result looks good prelim
Intravenous CD20 MS4A1
Ulocuplumab Human monoclonal antibody IgG4 Antineoplastic CLL phase I 2014
Phase I/II Waldenstrom macroglobulinemia 2025
Phase I/II AML 2021
Intravenous CXCR4 (CD184)
Urelumab
BMS-663513
Human monoclonal antibody IgG4κ Antineoplastic
CLL phase II 2020
Solid tumors phase II 2023
Intravenous Human receptor 4-1BB (CD137)
Urtoxazumab
TMA-15
Humanized monoclonal antibody IgG1κ Disease modifying EHEC animal studies Escherichia coli (EHEC) shiga toxin 2
Ustekinumab Stelara
FDA 2009
EU 2009
Human monoclonal antibody IgG1κ Disease modifying
Crohn disease
Plaque psoriasis
Psoriatic arthritis
Subcutaneous
Intravenous
p40 subunit of interleukin 12 (IL-12p40), IL-23
Utomilumab
PF-05082566
Human monoclonal antibody IgG2 Antineoplastic
Diffuse large B-cell lymphoma
Phase I 2021 phase II 2020
Breast phase II 2025
Intravenous 4-1BB (CD137)
Vadastuximab talirine
H2H12EC
Chimeric monoclonal antibody IgG1κ Antineoplastic
Acute myeloid leukemia phase II 2017 phase III 2017
MDS phase II 2017
Intravenous CD33
Vanalimab
Mitazalimab
Humanized monoclonal antibody IgG1λ Antineoplastic? No studies clinical trial or PubMed Immune checkpoint receptor, tumor necrosis receptor family CD40, (TNFRSF5)
Vandortuzumab vedotin Humanized monoclonal antibody Antineoplastic
Prostate cancer
STEAP1
Vantictumab
OMP-18R5
Human monoclonal antibody IgG2mab Antineoplastic
NSCLC, breast phase I 2017
Intravenous Frizzled receptor
Vanucizumab
RG-7221
RO5520985
Humanized monoclonal antibody IgG1κ bispecific ANG-2/VEGF-Amab Antineoplastic
Phase I 2018
Intravenous Angiopoietin 2/vascular endothelial growth factor A
Vapaliximab
BTT-1002
HUVAP
Chimeric monoclonal antibody IgG2κ No studies in PubMed or clinical trials Vascular adhesion protein AOC3 (VAP-1)
Varisacumab
GNR-011
R-84
Human monoclonal antibody IgG1κ No studies in PubMed or clinical trials VEGF-A
Varlilumab
CDX-1127
Human monoclonal antibody IgG1κ Antineoplastic
Solid tumors and hematologic malignancies
Phase I 2017, phase II 2019/20
Melanoma phase II 2018/21
Intravenous CD27
Vatelizumab
GBR500
SAR339658
Humanized monoclonal antibody IgG4 Disease modifying
UC phase II 2016
MS phase II 2016 withdrawn lack of efficacy
Α2β1 integrin I domain ITGA2 (CD49b)
Vedolizumab
LDP02
MLN02
Entyvio
FDA 2014
EU 2014
Humanized monoclonal antibody IgG1κ Disease modifying
Crohndisease
Ulcerativecolitis
In CD resolution extraintestinal manifestations
Intravenous Integrin α4β7
Selectively blocks trafficking of
Memory T cells to inflamed gut tissue by inhibiting a4b7-mucosal addressin cell adhesion molecule-1 (MAd-CAM-1) interaction
Veltuzumab
IMMU-106
HA20
Humanized monoclonal antibody IgG1κ Antineoplastic
Non-Hodgkin's lymphoma phase II 2013
ITP phase II 2016
Subcutaneous CD20
Vepalimomab Murine monoclonal antibody AOC3 (vascular adhesion protein-1)
Vesencumab
MNRP1685A
Human monoclonal antibody IgG1mab Antineoplastic
Solid malignancies
Phase I 2011 proteinuria
Neuropilin1 (NRP1)
Visilizumab Nuvion Humanized monoclonal antibody IgG2 Disease modifying
Prevent GVHD
Not effective in UC
CD3
Vobarilizumab Humanize monoclonal scFv Disease modifying inflammatory autoimmune diseases Nothing in PubMed IL6R
Volociximab
M200
Chimeric monoclonal antibody IgG4κ Antineoplastic
Solid tumors
NSCLC phase I/II 2010
Disease modifying phase I AMD terminated no results
Integrin α5β1
Vopratelimab
JTX-2011
Humanized monoclonal antibody IgG1κ Antineoplastic
Solid tumors phase II 2022
Intravenous Inducible T-cell costimulator (ICOS)
Vorsetuzumab mafodotin
H1F6
SGN-70
Humanized monoclonal antibody Antineoplastic
Phase I 2017
Intravenous CD70
Votumumab HumaSPECT
Diagnostic
EU 1998
Withdrawn from market 2003
Human monoclonal antibody Diagnostic
Human colon cancer imaging
Tumor antigen Cytokeratin tumor-associated antigen (CTAA16.88)
Vunakizumab
SHR-1314
Humanized monoclonal antibody IgG1 Disease modifying
Psoriasis phase II 2019
Subcutaneous
Nothing published
Interleukin 17 alpha
Xentuzumab
BI-836845
Humanized monoclonal antibody Antineoplastic
Breast, prostate, solid phase I 2019
Intravenous
No clinical studies published
Insulin-like growth factor (IGF1, IGF2)
XMAB-5574
Tafasitamab
MOR00208
FDA review possible 2019 Humanized monoclonal immunoglobulin fragment κ Fc Antineoplastic
Diffuse large B-cell lymphoma phase II 2015/18/19/22
phase III 2022
Intravenous CD19
Zalutumumab
2F8
HUMAX-EGFR
HuMax-EGFr
Suspended for commercialization
Human monoclonal antibody Antineoplastic
Squamous cell carcinoma of the head and neck phase II 2011
phase III 2016
Intravenous EGFR
Zanolimumab HuMax-CD4 (trade name) Humanized monoclonal antibody IgG1κ Antineoplastic CTCL
Phase II good results
Phase III suspended by company?
Intravenous CD4
Zenocutuzumab Humanized monoclonal antibodyIgG1 bispecific epidermal growth factor receptors her2,her3 Antineoplastic ERBB3, HER3
Ziralimumab Human monoclonal antibody IgM Disease modifying
immunosuppressive
No studies clinical trials or PubMed CD147 (basigin)
Zolbetuximab
IMAB362
Claudiximab
Chimeric monoclonal antibody IgG1κ ADCC enhance antibody Antineoplastic gastric cancer phase I, IIb
Phase III 2023
Gastrointestinal adenocarcinomas and pancreatic tumor
Intravenous Claudin protein (CLDN18.2)
Zolimomab aritox
H65-RTA
ZX-CD5
Orthozyme CD 5 plus Human monoclonal antibody IgG1 Disease modifying Not effective in preventing GVHD 1994 CD5

Auristatins are water-soluble dolastatin analogs of dolastatin 10. Dolastatin 10 belongs to dolastatin family and it can powerfully bind to tubulin, thus inhibiting polymerization mediated through the binding to the vinca alkaloid-binding domain, and causes cell to accumulate in metaphase arrest.

Ulcerative Colitis

Mabs being studied for UC but not yet approved by the FDA include bimekizumab, etrolizumab, golimumab, mirikizumab, ravagalimab, sacituzumab govitecan, ontamalimab, and vatelizumab. Refer to Table 16.1.

Autoimmune Diseases

Autoimmune diseases affect many organs and tissues including liver, gall bladder, pancreas (β islet cells in diabetes mellitus), nerve junctions (myasthenia gravis), thyroid, bone and joints, blood vessels, and multiorgan systems, systemic lupus erythematosus (SLE). Autoimmune arthritis is of multiple types including psoriatic, sclerosis, rheumatoid arthritis (RA), and SLE. Many of these diseases are mediated by antibody or cellular autoimmunity but ultimately appear to be secondary to an underlying abnormality in T-cell immune-regulatory control. These disease processes are historically controlled with antiinflammatory agents, immunosuppressive/immunomodulatory agents, or low-dose chemotherapy. Those with resultant hormone deficiencies are supplemented with hormones depleted by the disease process. It is hoped that passive antibody therapy will mitigate the sequelae of these inflammatory processes.

Plaque psoriasis/psoriatic arthritis

Psoriasis affects 2%–3% of the world population and is an inflammatory skin disease. Brodalumab (Siliz) is a human Mab (IgG2κ) with specificity to IL-17 receptor A (IL-17RA). It is FDA approved for treatment of plaque psoriasis, and its mechanism of action is by inhibiting IL-17A, IL-17F, IL-17C, IL-25, and IL-17A/F heterodimer cytokine-induced responses including release of proinflammatory cytokines. When compared to ustekinumab, response rates nearly doubled with brodalumab in phase II and phase III trials during induction and maintenance therapies.275, 276

Other therapies currently also approved or being studied for treatment of this disease include bermekimab (MABp1,T2-18C3, CA-18C3, Xilonix), bimekizumab, briakinumab, certolizumab pegol (Cimzia), etanercept (Enbrel), infliximab (Remicade, Inflectra, Remsima), itolizumab (Alzumab), adalimumab (Humira, Amjevita), ustekinumab (Stelara), secukinumab (AIN457, Cosentyx), guselkumab (Tremfya), tildrakizumab (MK-3222, SCH-900,222, Ilumya, Ilumetri), risankizumab (ABBV-066, BI-655,066), mirikizumab (LY3074828), namilumab (MT203), netakimab, and vunakizumab. Refer to Table 16.1.

Withdrawn from market or ineffective for treating psoriasis include efalizumab (Raptiva), fezakinumab, bleselumab, and teplizumab (MGA031, PRV-031, hOKT3g1(Ala-Ala)) Refer to Table 16.1.

Systemic juvenile idiopathic arthritis

Abatacept (Orencia) is a recombinant soluble fusion protein of the extracellular domain of human cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) linked to the modified Fc portion of human IgG1. Its mechanism of action is as selective costimulation modulator as it inhibits T lymphocyte activation by binding to CD80 and CD86, thereby blocking interaction with CD28. This interaction provides a costimulatory signal necessary for full activation of T lymphocytes. This medication is FDA approved for both juvenile idiopathic arthritis (JIA) and adult RA.277, 278, 279

Rheumatoid Arthritis

Certolizumab pegol alone or with methotrexate improves quality of life in RA and may cause disease remission and reduce joint damage.280

Ankylosing Spondylitis

Certolizumab pegolis is also approved for use with ankylosing spondylitis.

Systemic Lupus Erythematosus

Belimumab (Benlysta) is a human Mab (IgG1λ) that binds to B-cell activating factor and acts as a B-lymphocyte stimulator-specific inhibitor. It was approved by the FDA in 2011 for treatment of adult patients with active, autoantibody-positive SLE receiving standard therapy. This medication also decreases episodic frequency of lupus nephritis.281, 282, 283

Cardiovascular Disease

Despite marked improvement in survival from cardiovascular disease, this illness remains the number one cause of mortality in the US. This process causes injury to the endothelium of blood vessels of the heart secondary to toxins, accumulation of cholesterol, or chronic low-grade inflammation. Treatment has been preventive, primarily during actual injury or following injury. Therapies involve changes in behavior (diet, exercise, and cessation of tobacco use), pharmacologic to control contributing underlying illness (hypercholesterolemia, hypertension, diabetes type I and II), to diminish injury through thrombolytics, stents, vasodilators, supplemental oxygen, or to control sequelae of infarctions (cardiac dysfunction/failure). Passive antibody therapies are being tried to decrease the effects of some of the contributing factors of atherosclerotic plaque formation.

Abciximab (ReoPro) is a chimeric recombinant monoclonal fragment (IgG1 Fab’) with specificity to platelet glycoprotein IIb/IIIa receptor (CD41 7E3)/Intergrin α-IIb that prevents platelets from binding to fibrinogen. This Mab also prevents coagulation factor XIII from binding to platelets allowing stabilization of clots and are more easily lysed. The Fc portion of the antibody is removed to decrease thrombocytopenias. This antibody is used during high-risk coronary intervention to prevent clot formation and cardiac ischemia.284

Alirocumab (Praluent) is a human Mab (IgG1) with specificity to proprotein convertase subtilisin/kexin type 9. This medication is used to control cholesterol levels in patients at high risk for cardiovascular events and in patients with familial hypercholesterolemia who are not controlled by other agents.285, 286, 287

Evolocumab (Repatha) is a human Mab (IgG2λ) FDA approved for the treatment of hypercholesterolemia in patients with familial hypercholesterolemia or history of cardiovascular disease. This Mab has specificity to PCSK9. This medication reduced low-density lipoprotein (LDL) and cholesterol levels by 60% even after statin therapy. Hazard ratios for primary and secondary endpoints were less than one (∼0.80–0.85) with fewer cardiovascular-related death or infarction and stroke.288, 289

Under future watch is frovocimab (LY3015014) a humanized Mab (IgG4κ) with specificity to PCSK9 that completed phase I and II trials. There was up to 50% reduction in LDL cholesterol levels. Phase III studies have yet to be performed.290

An additional antibody is lodelcizumab a humanized Mab (IgG1κ); however, no studies were found in clinicaltrials.gov or in Pubmed searches.

Bococizumab is a humanized Mab (IgG2κ) that was in phase III trial, which was discontinued secondary to primary endpoints not being achieved.291

Neurologic Diseases

Besides autoimmune and malignant diseases of the neurologic system, there are also diseases of the central nervous system classified as degenerative. Such diseases include supranuclear palsy (SNP), Alzheimer's, and Parkinson's. Alzheimer's is likely the most common cause of dementia first described in 1907. This disease may be depicted as presenile or senile dementia and progresses at a similar rate no matter age of onset. This disease has a genetic predisposition causing it to occur in younger age groups. Histological changes include diffuse plaques (containing amyloid), neurofibrillary plaques, and neuronal loss especially in the hippocampus and temporal regions. Medical management may reverse some of the symptoms but does not prevent disease progression. Parkinson's is a mainly sporadic degenerative disease with a gradual progressive course mainly affecting motor function more than memory. It was first described in 1817. This is a disease of the substantia nigra characterized by loss of melanin containing nerve cells and eosinophilic intracytoplasmic inclusions. Aside from emotional support and physical therapy, medical therapy is used to decrease tremors including anticholinergic drugs for tremors at onset, beta blockers for intention tremors, and levodopa for postural imbalance and akinesia. Deep brain stimulation is also used to treat symptoms later on as disease progresses. SNP starts in the same age range as Parkinson's (middle to later in life) that was first described in 1963 with disturbances in gait and balance secondary to rigidity of trunk muscles. Loss of neurons and gliosis is seen in the midbrain. Medical treatment is relatively unsuccessful. Multiple sclerosis is a demyelinating disease most often seen in young adults. The clinical manifestations are diverse and the progression can be chronic, acute, or remitting and relapsing. Medications and therapeutic plasma exchange have been used to treat this debilitating disease with limited efficacy. Clinical trials are ongoing looking at Mab therapies for treatment of these four neurologic degenerative diseases.

Multiple Sclerosis

Alemtuzumab (Lemtrada) is a humanized Mab (IgG1κ) targeting CD52 that depletes lymphocytes (B and T cell) as reported earlier and is FDA approved for treatment of acute relapsing and remitting multiple sclerosis.292

Ocrelizumab (Ocrevus) is a humanized Mab (IgG1κ) with specificity to CD20 (a B-cell membrane protein). In phase II trials, there were decreases in brain lesions on imaging, and decrease rate of disability decline in primary progressive multiple sclerosis.293

Natalizumab (Tysabri) is a monoclonal IgG4κ humanized antibody with specificity to cell adhesion molecule (CD62L) that is FDA approved for relapsing multiple sclerosis.294, 295

The mabs to watch out for in the future and are in clinical trials include anifrolumab a human monoclonal antibody in phase I trials; elezanumab is a human Mab (IgG1λ) with specificity to repulsive guidance molecule family member-A that is in phase II trials to be completed 2021; and finally inebilizumab (MEDI-551) is a humanized monoclonal antibody (IgG2κ) with specificity to CD19 (a B-cell lymphocyte protein). This Mab mechanism of action is via ADCC and has completed phase I trials with good safety profile and response in decreasing lesions seen on contrast enhanced magnetic resonance imaging. Otilimabis (MOR103) is a human Mab (IgG1λ) completing phase I studies with good safety profile that targets granulocyte-macrophage colony-stimulating factor. Ublituximab is in phase II clinical studies to be completed in 2019, and phase III studies are scheduled to be completed in 2021. This Mab is a chimeric Mab (IgG1κ) with specificity to CD20 MS2A1.296, 297, 298, 299, 300

Additional Mab have serious adverse effects such as daclizumab a humanized monoclonal (IgG1κ) with specificity to (CD25 {IL-2Rα}); or are ineffective as is opicinumab a human Mab IgG1 with specificity to Leucine-rich repeat and immunoglobulin domain containing neurite outgrowth inhibitor receptor interacting protein-1 which in a phase II trial was no more beneficial than placebo in treating optic neuritis in multiple sclerosis patients.301, 302

Alzheimer's Disease

Aducanumab is a human Mab IgG1 with specificity to β-amyloid (N-terminus 3–6) soluble oligomers and insoluble fibers. Phase III clinical trials are ongoing since 2015.

BAN-2401 is a humanized Mab IgG1 with specificity to β-amyloid fibrillary and soluble β amyloid and is in phase IIb clinical studies since 2013.

Gosuranemab (BIIB092, IPN-007) is a humanized Mab IgG4κ with specificity to the tau protein and is in clinical trials to treat Alzheimer's disease scheduled to be completed in 2021. Gosuranemab is also in phase I studies to treat progressive suranuclear palsy and will be completed in 2020.

Crenezumab (RG7412, MABT5102A) is a humanized Mab IgG4 with specificity to 1–40 β-amyloid and is on phase III studies scheduled to be completed in 2021 and 2022.

Gantenerumab (R04909832, R1450) is a human Mab IgG1κ with targets β-amyloid. This Mab on initial phase III studies was found to be ineffective. Ongoing phase II/III trials are currently in place at higher dosing in a clinical population of people with autosomal dominant form of Alzheimer's disease.

Solanezumab (LY2062430) is a humanized Mab IgG1 with specificity to beta amyloid. Initial phase III trials discontinued for lack of efficacy in preventing Alzheimer's disease. Ongoing phase III trials are now in place for secondary prevention of this disease and will be completed in 2021 and 2022.

Mab antibodies studied and were ineffective include bapineuzumab, gantenerumab (R04909832, R1450), and ponezumab (RN1219, PF-04,360,365).

Parkinson's Disease

Prasinezumab (PRX002, RG7935, RO7046015) is a humanized Mab IgG1κ with specificity to α-synuclein. This Mab is in phase II clinical trials to treat Parkinson's and will be completed in 2021.

Allergic Diseases

Allergic reactions develop because of immunologic stimulation of IgE antibodies followed by their interaction with allergens and mast cells. Effects can be local (dermatitis) or systemic (respiratory, cardiovascular, and gastrointestinal). Treatment is either avoidance of the allergens or supportive therapy in acute allergic reactions including pharmacologic treatment with type 1 and 2 histamine blockers, glucocorticosteroids, and if life-threatening epinephrine. Passive antibody therapies are being studied and approved to curtail severe reactions.

Asthma

Asthma affects 24 million individuals in the US, and up to 10% of asthma patients have severe disease that may be uncontrolled despite high doses of standard-of-care asthma medications requiring additional use of chronic oral corticosteroids. Benralizumab (Fensenra) is a humanized Mab (IgG1κ) with specificity to CD125 (IL-5Rα). This Mab is approved to treat severe asthma of the eosinophilic subtype in ages 12 and older. Its mechanism of action is to decrease the number of eosinophils via ADCC. Basophils are also depleted.303

Atopic Dermatitis

Dupilumab (Dupixent) is a human monoclonal gG4 antibody with specificity to interleukin-4 receptor subunit-alpha (IL-4Rα) that is approved to treat severe atopic dermatitis in adults.304

Coagulopathy and Other Benign Hematologic Diseases

Coagulopathies are usually either autoimmune or genetic. In factor VIII deficiency, recombinant factor VIII is used to replace lack of this protein. However, patients may develop antibodies to factor VIII leading to high titers of inhibitors. Furthermore, patients without deficiency may also develop autoantibodies to factor VIII de novo leading to coagulopathies. Other factor combinations as well as recombinant active factors have been created to overcome these inhibitory antibodies. Mabs with bispecific binding are also being researched as another avenue for treatment.

ITP can lead to critical low platelet levels increasing risk for severe bleeding. ITP can occur in both adult and pediatric settings as it is considered an autoimmune disease. Typically, this is treated with steroids and IVIG. In addition, as mentioned earlier, RhD+ patients have benefitted from polyclonal medications directed against the D antigen. Recently, Mab to treat this disease have been developed and will be discussed next.

Thrombotic thrombocytopenic purpura (TTP) is a blood disorder that does not lead to bleeding but to development of diffuse thrombi in small blood vessels. More often, this disorder is secondary to an autoimmune inhibitory antibody to the disintegrin and metalloproteinase with thrombospondin type 1 motif member-13 (ADAMTS-13), known as acquired TTP. Inhibiting this zinc containing metalloprotease leads to lack of cleavage of large multimers of von Willebrand Factor (vWF). The large vWF multimers then more easily bind to platelets resulting in platelet clots in small blood vessels. More rarely, this disorder is secondary to an inherited deficiency of ADAMTS-13. This patient population with congenital deficiency is managed with transfusion of FFP to replace the deficient enzyme. Acquired TTP is typically treated with therapeutic plasma exchange (TPE). This treatment modality removes the inhibitory antibody and ultralarge vWF multimers. Similarly, TPE will replete the missing enzyme. Immunosuppressive agents may be added if only TPE is not effective. A Mab preventing interaction of vWF and platelets was recently approved for use in treating this disorder.305, 306 Caplacizumab-yhdp (Cablivi) is a humanized single-variable-domain immunoglobulin (Nanobody) that inhibits the interaction between ultralarge vWF multimers and platelets and is directed against vWF. It induces a faster response to therapy with TPE and decreases relapse with continued use during TPE. This medication is then used post-TPE treatment until immunological evidence of disease is controlled to prevent relapse.305, 307, 308 This medication was FDA approved for use in TTP in 2019.

Atypical hemolytic uremic syndrome (aHUS) is a disorder of the complement system due to uncontrolled activation. This disorder presents with thrombocytopenia, thrombi, and renal dysfunction. Historically, this illness was treated with TPE; however, end-stage renal failure occurred in 30% of patients and about 65% mortality in subsequent relapses with increasing incidence of renal failure. There are now two monoclonal antibodies approved for the treatment of aHUS. Refer to Table 16.1.

Sickle Pain Crisis

In sickle cell disease, one of the frequent complications is pain crises. This is usually treated with analgesics, oxygen, hydration, and transfusions (simple or exchange). Monoclonal antibodies are being developed to treat pain crises in sickle cell patients in both adult and pediatric populations. Crizanlizumab is a humanized Mab (IgG2κ) with specificity to selectin P. One phase II trial was completed in 2016 and three additional phase II studies will be completed between 2021 and 27 to treat vasoocclusive pain crisis. This medication may be under FDA review as early as 2019.309, 310

Infections

Antimicrobials have historically been developed against a variety of viral, bacterial, fungal, and parasitic infections. These pharmaceuticals target differences from human cells of these particular organisms such as cell wall or membrane structure, genetic make-up, transcription/translation of genetic material, or metabolic pathways. Often organisms develop resistance to entire categories of these medications. Earlier in the chapter, passive polyclonal antibodies were discussed in the treatment of some of these infectious agents and we will now discuss research in monoclonal therapies to pathogenic microorganisms.

Clostridium difficile

Enterocolitis from Clostridium difficile is a community or hospital acquired infection increasing morbidity and mortality in those that acquire it. Treatment is supportive or with fecal transplants or antibiotics. Bezlotoxumab (Zinplava) is a human Mab (IgG1) with specificity to Clostridium difficile's B toxin. It is used to treat pseudomembranous colitis and prevent C. difficile reinfection.311, 312

Actoxumab, a monoclonal antibody against C. difficile toxin A, has shown not to be clinically significant.

Respiratory Syncytial Virus

Respiratory syncytial virus (RSV) infects almost all children by 2 years old and poses extra risk in preterm infants. Supportive therapy, RSV-IG or IVIG, and antiviral therapy have been used to mitigate the sequela of this infection with optimal response yet to be seen. No vaccines have yet to be developed for this infection. Recently, monoclonal antibodies have been FDA approved or are undergoing pre/clinical trials to treat this infectious process and include palivizumab, Nirsevimab (MEDI8897), TRL3d3 (3D3), and ALX-0171.313, 314

Not beneficial or safe in use for RSV: motavizumab, Suptavumab (REGN2222, SAR438584).

Influenza virus

Influenza is a worldwide respiratory infectious problem with cyclic epidemics yearly. Supportive therapy, yearly vaccinations, and antivirals are used to decrease the morbidity and mortality caused by this sometimes virulent pathogen. Both polyclonal and monoclonal therapies are being evaluated to better treat these infections. Mabs in pre/clinical trials include diridavumab (CR6262), firivumab, gedivumab (RG7745, RO6876802), lesofavumab (RG70026), and Navivumab (CT-P27).

Rabies

Rabies is a devastating viral infection with swift mortality if not treated quickly after initial exposure. Vaccines usually react too slowly and have to be combined with polyclonal IVIG infusions. Monoclonal therapy was previously studied but usually the virus mutates quickly and the infection is not controlled. More recently, in clinical trials, cocktails of Mabs are being tried to more closely mimic the benefits of polyclonal therapies. These Mabs include foravirumab, rafivirumab (CR57), and Rmab.

Hepatitis B virus

HBV is one of if not the most common infections in the world. Even though antivirals are available and effective, only recently they have they been widely used in the infant population and not just “high”-risk individuals. Mabs to treat this infection that are being investigated include libivirumab. Mab that is not found to be effective is tuvirumab.

Ebola

Ebola is a relatively rare but devastating hemorrhagic infection. Most care is supportive with various studies being performed to prevent/mitigate this disease. Vaccines are under development as well as passive polyclonal therapies. Mab therapies being developed or studied include porgaviximab (C2G4), cosfroviximab, and larcaviximab.

For these and other bacterial, fungal, and viral antiinfectious agents, information may be found in Table 16.1.

Immunomodulation

In solid organ transplants, cellular or humoral immunity can develop against the transplant leading to acute or chronic rejection. An additional complication with these and stem cell transplants is severe GVHD. In the past, these transplant complications were treated with high-dose glucocorticosteroids, immunosuppressive medication, chemotherapeutic agents, IVIG, or T-cell lymphocytic specific immunoglobulins. Recently, Mabs have been added to this armamentarium to better control these adverse reactions to transplantations.

Basiliximab (Simulect) is a chimeric Mab (IgG1κ) with specificity to CD25 IL-2α. The only FDA-approved indication for this medication is prophylaxis of acute rejection in renal transplant patients. There are multiple ongoing studies of this biological for other organ transplants including liver, lung, and heart as well as for inflammatory/immunologic diseases such as GVHD following stem cell transplantation, ulcerative colitis, and uveitis.315, 316, 317, 318, 319

Belatacept (Nulojix) is a soluble fusion protein consisting of the modified extracellular domain of CTLA-4 fused to the Fc domain of a recombinant human Mab IgG1. This Mab selectively inhibits T-cell activation through costimulation blockade binding to both CD80 and CD86 while blocking CD28 via tighter binding than its parent antibody abatacept. Refer to Table 16.1.

Metabolic Syndromes

Hypercholesterolemia is associated with increased risk for cardiovascular disease/atherosclerosis secondary to inherited or dietary etiologies. Diet and exercise are used to treat mild forms of these disorders. Medications such as nicotinic acid, fibrates, bile acid binding resins, and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors are used for more severe forms of these disorders. Phase III studies have been completed with monoclonal antibodies for patients' refractory to the previously mentioned forms of therapy.

Hypophosphatemia

Burosumab (KRN23, Crysvita) is a human Mab IgG1κ with specificity to phosphaturic hormone fibroblast growth factor 23 (FGF 23). This hormone is a regulator of phosphate and vitamin D homeostasis. FGF23 inhibits the enzyme CYP27B1 and stimulates CYP24A1, thereby reducing circulating levels of 1,25-dihydroxyvitamin D (1,25(OH)2D), the active metabolite of vitamin D. This medication is FDA approved for the treatment of X-linked hypophosphatemic rickets.320, 321

Osteoporosis

Denosumab (Prolia) is an FDA-approved human Mab (IgG2) that is a receptor activator of nuclear factor κB ligand that inhibits development and activity of osteoclasts. As Prolia, this medication is used to prevent or treat osteoporosis in women.322, 323, 324 This medication under the trade name Xgeva is also used to prevent skeletal-related events in adults with bone metastasis from breast, prostate cancers, and multiple myeloma.325, 326

Endocrine Disorders

Diabetes may be classified as primary or secondary. In this chapter, we will be mainly interested in both insulin-dependent (Type I) and insulin-independent types (Type II). Type I diabetes mellitus is generally secondary to loss of β cells in the islets of Langerhans and subsequent loss of insulin production. Type II typically is secondary to decreased sensitivity to the effects of insulin. In type I, insulin is replaced exogenously depending on glucose levels. In type II, medications are given to stimulate islet cells to produce more insulin. Mabs are being developed to potentially mitigate the autoimmune process leading to Type I diabetes mellitus or the sequela of renal failure often seen with this disease. For type II, Mabs are being investigated to potentially decrease body mass index and thus decrease disease severity. Refer to Table 16.1.

Other Clinical Disorders

Age-related macular degeneration (AMD) is the leading irreversible cause of visual loss affecting the elderly. Two forms include a dry form with deposits in the macula or a wet form involving abnormal growth of blood vessels. The wet form, even though less frequent, is associated with more severe visual acuity loss. Antiangiogenesic drugs or laser treatments are used to slow the progression or even partially reverse visual loss. Some trials have been completed while others are ongoing using Mab to treat the wet form of AMD. Brolucizumab was found as good as if not better than aflibercept in a phase III clinical trial.327

Cryopyrin-associated periodic syndromes (including familial cold auto-inflammatory syndrome and Muckle-Wells syndrome); tumor necrosis factor receptor-associated periodic syndrome (TRAPS); hyperimmunoglobulin D Syndrome (HIDS)/mevalonate kinase deficiency and familial Mediterranean fever (FMF) may also respond to canakinumab.328

Potential Future Uses of Monoclonal Antibodies and Their Targets

Passive antibody therapy continues to be useful clinically whether polyclonal or monoclonal therapy is implemented. Increased utilization of the classic polyclonal antibody preparations continue especially in the realm of infections. In the past 3 years, monoclonal therapy has evolved and revolutionized treatment in many areas. As targets are identified to modify disease pathology no matter its genre we continue to get a better handle on morbidity and mortality. We are learning that not only is the target important put the portion of the target mediating the effect we intend to modify is also important. Importantly, modification of antibodies to be more compatible with the immune system while decreasing rapidity of clearance also allows for more consistent therapy. There are also many targets yet to be discovered or only now being developed as in the canonical wingless/integrated (WNT) signaling. This receptor family is important in a multitude of diseases not limited to: hereditary colorectal cancer, various types of sporadic cancers, intellectual disability syndrome, Alzheimer's disease, bipolar disorder, bone diseases, and vascular diseases. One monoclonal antibody rosmantuzumab (OMP-131R10), a humanized Mab (IgG1κ), is in phase I trials to treat colorectal cancer.329, 330 Other disease processes have yet to find their optimal therapy (Alzheimer's) or are advancing to fuller therapeutic benefit. The future is wide open for this newer class of pharmaceuticals as they continue to develop to full fruition.

References

  • 1.Sangstat Medical Corporation . December 1998. Thymoglobulin; (Anti-thymocyte Globulin [rabbit]) Prescribing Information. Menlo Park, CA. [Google Scholar]
  • 2.Ormrod D., Jarvis B. Antithymocyte globulin (rabbit): a review of the use of Thymoglobulin in the prevention and treatment of acute renal allograft rejection. BioDrugs. 2000;14:255–273. [Google Scholar]
  • 3.Pharmacia. Atgam Prescribing Information. June 2000. Kalamazoo, MI. [Google Scholar]
  • 4.The Upjohn Company . November 1981. Drug Reference: Atgam. Kalamazoo, MI. [Google Scholar]
  • 5.Cosimi A.B. The clinical value of antilymphocyte antibodies. Transplant Proc. 1981;13:462–468. [PubMed] [Google Scholar]
  • 6.Cosimi A.B. The clinical usefulness of antilymphocyte antibodies. Transplant Proc. 1983;15:583–589. [Google Scholar]
  • 7.Cho S.I., Bradley J.W., Carpenter C.B. Antithymocyte globulin, pretransplant blood transfusion, and tissue typing in cadaver kidney transplantation. Am J Surg. 1983;145:464–471. doi: 10.1016/0002-9610(83)90041-7. [DOI] [PubMed] [Google Scholar]
  • 8.Nelson P.W., Cosimi A.B., Delmonico F.L. Antithymocyte globulin as the primary treatment for renal allograft rejection. Transplantation. 1983;36:587–589. [PubMed] [Google Scholar]
  • 9.Nowygrod R., Appel G., Hardy M.A. Use of ATG for reversal of acute allograft rejection. Transplant Proc. 1981;13:469–472. [PubMed] [Google Scholar]
  • 10.Hardy M.A., Nowygrod R., Elberg A. Use of ATG in treatment of steroid-resistant rejection. Transplantation. 1980;29:162–164. doi: 10.1097/00007890-198002000-00015. [DOI] [PubMed] [Google Scholar]
  • 11.Simonian S.J., Lyons P., Chvala R. Reversal of acute cadaveric renal allograft rejection with added ATG treatment. Transplant Proc. 1983;15:604–607. [Google Scholar]
  • 12.Gaber A.O., First M.R., Tesi R.J. Results of the double-blind, randomized, multicenter, phase III clinical trial of Thymoglobulin versus Atgam in the treatment of acute graft rejection episodes after renal transplantation. Transplantation. 1998;66:29–37. doi: 10.1097/00007890-199807150-00005. [DOI] [PubMed] [Google Scholar]
  • 13.Brennan D.C., Flavin K., Lowell J.A. A randomized, double-blinded comparison of Thymoglobulin versus Atgam for induction immunosuppressive therapy in adult renal transplant recipients. Transplantation. 1999;67:1011–1018. doi: 10.1097/00007890-199904150-00013. [DOI] [PubMed] [Google Scholar]
  • 14.Lance E.M. Mode of action of antilymphocyte serum. Fed Proc. 1970;29:209–211. [PubMed] [Google Scholar]
  • 15.Martin W.J., Miller J.F.A.P. Site of action of antilymphocyte globulin. Lancet. 1967;2:1285–1287. doi: 10.1016/s0140-6736(67)90395-9. [DOI] [PubMed] [Google Scholar]
  • 16.Levey R.H., Medawar P.B. Nature and mode of action of antilymphocytic antiserum. Proc Natl Acad Sci USA. 1966;56:1130–1137. doi: 10.1073/pnas.56.4.1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wohlman M.H., Toledo-Pereyra L.H., Zeichner W.D. The immunosuppressive properties of antilymphocyte serum preparations: a current review. Dial Transplant. 1981;10:19–28. [Google Scholar]
  • 18.Zimmerman B., Tsui F. Immunosuppressive antilymphocyte serum: different subpopulations of T lymphocytes are influenced at different doses of antilymphocyte serum. Transplantation. 1979;28:323–328. doi: 10.1097/00007890-197910000-00012. [DOI] [PubMed] [Google Scholar]
  • 19.Bach J.F. Mechanism and significance of rosette inhibition by antilymphocyte serum. In: Bach J.F., Dormont J., Eyquem A., editors. Vol. 16. S Karger; New York: 1970. pp. 189–198. (International Symposium on Antilymphocyte Serum; Symposium Series on Immunobiology Standardization). [Google Scholar]
  • 20.Pirofsky B., Beaulieu R., Bardana E.J. Antithymocyte antiserum effects in man. Am J Med. 1974;56:290–296. doi: 10.1016/0002-9343(74)90610-x. [DOI] [PubMed] [Google Scholar]
  • 21.Greco B., Bielory L., Stephany D. Antithymocyte globulin reacts with many normal human cell types. Blood. 1983;62:1047–1054. [PubMed] [Google Scholar]
  • 22.Bonifazi F, Solano C, Wolschke C et al. GVHD prophylaxis plus ATLG after myeloablative allogeneic haemopoietic peripheral blood stem-cell transplantation from HLA-identical siblings in patients with acute leukaemia in remission: final results of quality of life and long-term outcome analysis of a phase 3 randomised study. The Lancet. Hematology, ISSN: 2352-3026, Vol: 6, Issue: 2, Page: e89-e99. 10.1016/S2352-3026(18)30214-X [DOI] [PubMed]
  • 23.Gale R.P., moderator Aplastic anemia: biology and treatment. Ann Intern Med. 1981;95:477–494. doi: 10.7326/0003-4819-95-4-477. Champlin RE. Treatment of aplastic anemia, pp. 480–483. In: [DOI] [PubMed] [Google Scholar]
  • 24.Champlin R., Ho W., Gale R.P. Antithymocyte globulin treatment in patients with aplastic anemia. N Engl J Med. 1983;308:113–118. doi: 10.1056/NEJM198301203080301. [DOI] [PubMed] [Google Scholar]
  • 25.Cheeseman S.H., Rubin R.H., Stewart J.A. Controlled clinical trial of prophylactic human-leukocyte interferon in renal transplantation: effects on cytomegalovirus and herpes simplex virus infections. N Engl J Med. 1979;300:1345–1349. doi: 10.1056/NEJM197906143002401. [DOI] [PubMed] [Google Scholar]
  • 26.Cheeseman S.H., Henle W., Rubin R.H. Epstein-Barr virus infection in renal transplant recipients: effects of antithymocyte globulin and interferon. Ann Intern Med. 1980;93(Part 1):39–42. doi: 10.7326/0003-4819-93-1-39. [DOI] [PubMed] [Google Scholar]
  • 27.Diethelm A.G., Aldrete J.S., Shaw J.F. Clinical evaluation of equine antithymocyte globulin in recipients of renal allografts: analysis of survival, renal function, rejection, histocompatibility, and complications. Ann Surg. 1974;180:20–28. doi: 10.1097/00000658-197407000-00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Talecris Biotherapeutics . Research Triangle Park; NC: May 2008. HyperTET S/D (Tetanus Immune Globulin [human]) Solvent/detergent Treated Prescribing Information. [Google Scholar]
  • 29.Centers for Disease Control and Prevention General recommendations on immunization: recommendations of the advisory committee on immunization practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2006;55(RR-15):1–48. [PubMed] [Google Scholar]
  • 30.Murphy T.V., Slade B.A., Broder K.R. Prevention of pertussis, tetanus, and diphtheria among pregnant and postpartum women and their infants recommendations of the Advisory Committee on Immunization Practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2008;57:1–51. [PubMed] [Google Scholar]
  • 31.Centers for Disease Control and Prevention . 11th ed. Public Health Foundation; Washington, DC: 2009. Epidemiology and Prevention of Vaccine-Preventable Diseases. [Google Scholar]
  • 32.CSL Behring . November 2010. CytoGam (Cytomegalovirus Immune Globulin Intravenous [human][CMV-IG]) Liquid Formulation Solvent Detergent Treated Prescribing Information. Kankakee, IL. [Google Scholar]
  • 33.Snydman D.R., McIver J., Leszczynski J. A pilot trial of a novel cytomegalovirus immune globulin in renal transplant recipients. Transplantation. 1984;38:553–557. doi: 10.1097/00007890-198411000-00026. [DOI] [PubMed] [Google Scholar]
  • 34.Snydman D.R., Werner B.G., Heinze-Lacey B. Use of cytomegalovirus immune globulin to prevent cytomegalovirus disease in renal-transplant recipients. N Engl J Med. 1987;317:1049–1054. doi: 10.1056/NEJM198710223171703. [DOI] [PubMed] [Google Scholar]
  • 35.Snydman D.R. Prevention of cytomegalovirus-associated diseases with immunoglobulin. Transplant Proc. 1991;23(Suppl 3):131–135. [PubMed] [Google Scholar]
  • 36.Snydman D.R., Werner B.G., Tilney N.L. Final analysis of primary cytomegalovirus disease prevention in renal transplant recipients with a cytomegalovirus-immune globulin: comparison of the randomized and open-label trials. Transplant Proc. 1991;23:1357–1360. [PubMed] [Google Scholar]
  • 37.Werner B.G., Snydman D.R., Freeman R. Cytomegalovirus immune globulin for the prevention of primary CMV disease in renal transplant patients: analysis of usage under treatment IND status. Transplant Proc. 1993;25:1441–1443. [PubMed] [Google Scholar]
  • 38.Ho M. Cytomegalovirus. In: Mandell G.L., Bennett J.E., Dolin R., editors. Mandell, Douglas and Bennett's Principles and Practice of Infectious Diseases. 4th ed. Churchill Livingstone; New York: 1995. pp. 1351–1364. [Google Scholar]
  • 39.Dickinson B.I., Gora-Harper M.L., McCraney S.A. Studies evaluating high-dose acyclovir, intravenous immune globulin, and cytomegalovirus hyperimmunoglobulin for prophylaxis against cytomegalovirus in kidney transplant recipients. Ann Pharmacother. 1996;30:1452–1464. doi: 10.1177/106002809603001215. [DOI] [PubMed] [Google Scholar]
  • 40.Snydman D.R. Cytomegalovirus immunoglobulins in the prevention and treatment of cytomegalovirus disease. Clin Infect Dis. 1990;12(Suppl 7):S839–S848. doi: 10.1093/clinids/12.supplement_7.s839. [DOI] [PubMed] [Google Scholar]
  • 41.Patel R., Snydman D.R., Rubin R.H. Cytomegalovirus prophylaxis in solid organ transplant recipients. Transplantation. 1996;61:1279–1289. doi: 10.1097/00007890-199605150-00001. [DOI] [PubMed] [Google Scholar]
  • 42.Meyers J.D. Prevention of cytomegalovirus infection after marrow transplantation. Rev Infect Dis. 1989;11(Suppl 7):S1691–S1705. doi: 10.1093/clinids/11.supplement_7.s1691. ([PubMed]) [DOI] [PubMed] [Google Scholar]
  • 43.Winston D.J., Ho W.G., Champlin R.E. Cytomegalovirus infections after allogeneic bone marrow transplantation. Clin Infect Dis. 1990;12(Suppl 7):S776–S787. doi: 10.1093/clinids/12.supplement_7.s776. [DOI] [PubMed] [Google Scholar]
  • 44.Valantine H.A. Prevention and treatment of cytomegalovirus disease in thoracic organ transplant patients: evidence for a beneficial effect of hyperimmune globulin. Transplant Proc. 1995;27(Suppl 1):49–57. [PubMed] [Google Scholar]
  • 45.Grundy J.E. Virologic and pathogenetic aspects of cytomegalovirus infection. Clin Infect Dis. 1990;12(Suppl 7):S711–S719. doi: 10.1093/clinids/12.supplement_7.s711. [DOI] [PubMed] [Google Scholar]
  • 46.Bass E.B., Powe N.R., Goodman S.N. Efficacy of immune globulin in preventing complications of bone marrow transplantation: a meta-analysis. Bone Marrow Transplant. 1993;12:273–282. [PubMed] [Google Scholar]
  • 47.Taylor-Wiedeman J., Sissons J.G., Borysiewicz L.K., Sinclair J.H. Monocytes are a major site of persistence of human cytomegalovirus in peripheral blood mononuclear cells. J Gen Virol. 1991;72:2059–2064. doi: 10.1099/0022-1317-72-9-2059. [DOI] [PubMed] [Google Scholar]
  • 48.Tsinontides A.C., Bechtel T.P. Cytomegalovirus prophylaxis and treatment following bone marrow transplantation. Ann Pharmacother. 1996;30:1277–1290. doi: 10.1177/106002809603001113. [DOI] [PubMed] [Google Scholar]
  • 49.Zamora M.R., Fullerton D.A., Campbell D.N. Use of cytomegalovirus (CMV) hyperimmune globulin for prevention of CMV disease in CMV-seropositive lung transplant recipients. Transplant Proc. 1994;26(Suppl 1):49–51. [PubMed] [Google Scholar]
  • 50.Snydman D.R., Werner B.G., Dougherty N.N. A further analysis of the use of cytomegalovirus immune globulin in orthotopic liver transplant patients at risk for primary infection. Transplant Proc. 1994;26(Suppl 1):23–27. [PubMed] [Google Scholar]
  • 51.Aguado J.M., Gomez-Sanchez M.A., Lumbreras C. Prospective randomized trial of efficacy of ganciclovir versus that of anti-cytomegalovirus (CMV) immunoglobulin to prevent CMV-seropositive heart transplant recipients treated with OKT3. Antimicrob Agents Chemother. 1995;39:1643–1645. doi: 10.1128/aac.39.7.1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Merck & Co . February 2014. Inc. Antivenin (Latrodectus Mactans) (Black Widow Spider Antivenin) Equine Origin Prescribing Information. Whitehouse Station, NJ. [Google Scholar]
  • 53.Clark R.F., Wethern-Kestner S., Vance M.V. Clinical presentation and treatment of black widow spider envenomation: a review of 163 cases. Ann Emerg Med. 1992;21:782–787. doi: 10.1016/s0196-0644(05)81021-2. [DOI] [PubMed] [Google Scholar]
  • 54.Wyeth Laboratories Inc . August 2001. Antivenin (Micrurus fulvius) (Equine Origin) (North American Coral Snake Antivenin) Prescribing Information. Marietta, PA. [Google Scholar]
  • 55.The United States Pharmacopeia, 25th Rev, and the National Formulary. 20th ed. The United States Pharmacopeial Convention, Inc; Rockville, MD: 2002. p. 158. [Google Scholar]
  • 56.Protherics Inc Crofab . 2010 Sep. (Crotalidae Polyvalent Immune Fab [ovine]) Prescribing Information. Brentwood, TN. [Google Scholar]
  • 57.Parrish H.M. Bites by coral snakes: report of 11 representative cases. Am J Med Sci. 1967;253:561. [PubMed] [Google Scholar]
  • 58.Russell F.E., editor. Snake Venom Poisoning. JB Lippincott Company; Philadelphia: 1980. Identification and distribution of North American venomous snakes; pp. 45–86. [Google Scholar]
  • 59.Roze J.A. New world coral snakes (Elapidae): a taxonomic and biologic summary. Mem Inst Butantan, Sao Paulo. 1982;46:305–338. [Google Scholar]
  • 60.Anon Treatment of snakebite in the USA. Med Lett Drugs Ther. 1982;24:87–89. [PubMed] [Google Scholar]
  • 61.Clark R.F., McKinney P.E., Chase P.B. Immediate and delayed allergic reactions to Crotalidae polyvalent immune Fab (ovine) antivenom. Ann Emerg Med. 2002;39:671–676. doi: 10.1067/mem.2002.123134. [DOI] [PubMed] [Google Scholar]
  • 62.Lavonas E.J., Ruha A.M., Banner W. Unified treatment algorithm for the management of crotaline snakebite in the United States: results of an evidence-informed consensus workshop. BMC Emerg Med. 2011;11:2. doi: 10.1186/1471-227X-11-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.England PH. MERS-CoV: clinical decision making support for treatment. https://www.gov.uk/government/publications/mers-covclinical-decision-making-support-for-treatment (accessed Nov 8, 2016).
  • 64.Luke T.C., Kilbane E.M., Jackson J.L., Hoffman S.L. Meta-analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment? Ann Intern Med. 2006;145:599–609. doi: 10.7326/0003-4819-145-8-200610170-00139. [DOI] [PubMed] [Google Scholar]
  • 65.Hung I.F., To K.K., lee C.K. Convalescent plasma treatment reduced mortality in patients with severe pandemic influenza A (H1N1) 2009 virus infection. Clin Infect Dis. 2011;52:447–456. doi: 10.1093/cid/ciq106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gulland A. First Ebola treatment is approved by WHO. BMJ. 2014;349:g5539. doi: 10.1136/bmj.g5539. [DOI] [PubMed] [Google Scholar]
  • 67.Mupapa K., Massamba M., Kibadi K. Treatment of Ebola hemorrhagic fever with blood transfusions from convalescent patients. International Scientific and Technical Committee. J Infect Dis. 1999;179:S18–S23. doi: 10.1086/514298. [DOI] [PubMed] [Google Scholar]
  • 68.Klein H.G. Should blood be an essential medicine? N Engl J Med. 2013;368:199–201. doi: 10.1056/NEJMp1213134. [DOI] [PubMed] [Google Scholar]
  • 69.Ala F., Allain J.P., Bates I. External financial aid to blood transfusion services in sub-Saharan Africa: a need for reflection. PLoS Med. 2012;9:e1001309. doi: 10.1371/journal.pmed.1001309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Burnouf T., Emmanuel J., Mbanya D. Ebola: a call for blood transfusion strategy in sub-Saharan Africa. Lancet. 2014;384:1347–1348. doi: 10.1016/S0140-6736(14)61693-7. [DOI] [PubMed] [Google Scholar]
  • 71.GlaxoSmithKline. Digibind (Digoxin Immune Fab [ovine]) Prescribing Information. Research Triangle Park; NC: September 2003. [Google Scholar]
  • 72.Smith T.W., Haber E., Yeatman L. Reversal of advanced digoxin intoxication with Fab fragments of digoxin-specific antibodies. N Engl J Med. 1976;294:797–800. doi: 10.1056/NEJM197604082941501. [DOI] [PubMed] [Google Scholar]
  • 73.Smith T.W., Butler V.P., Haber E. Treatment of life-threatening digitalis intoxication with digoxin-specific Fab antibody fragments. N Engl J Med. 1982;307:1357–1362. doi: 10.1056/NEJM198211253072201. [DOI] [PubMed] [Google Scholar]
  • 74.Cole P.L., Smith T.W. Use of digoxin-specific Fab fragments in the treatment of digitalis intoxication. Drug Intell Clin Pharm. 1986;20:267–270. doi: 10.1177/106002808602000403. [DOI] [PubMed] [Google Scholar]
  • 75.Smith T.W., Butler V.P., Haber E. Cardiac glycoside-specific antibodies in the treatment of digitalis intoxication. In: Haber E., Krause R.M., editors. Antibodies in Human Diagnosis and Therapy. Raven Press; New York: 1977. pp. 365–389. [Google Scholar]
  • 76.Wenger T.L., Butler V.P., Haber E. Treatment of 63 severely digitalis-toxic patients with digoxin-specific antibody fragments. J Am Coll Cardiol. 1985;5:118–123A. doi: 10.1016/s0735-1097(85)80471-x. [DOI] [PubMed] [Google Scholar]
  • 77.Larbig D., Raff U., Haasis R. Reversal of digitalis effects by specific antibodies. Pharmacology. 1979;18:1–8. doi: 10.1159/000137223. [DOI] [PubMed] [Google Scholar]
  • 78.Smolarz A., Roesch E., Lenz E. Digoxin specific antibody (Fab) fragments in 34 cases of severe digitalis intoxication. J Toxicol Clin Toxicol. 1985;23:327–340. doi: 10.3109/15563658508990641. [DOI] [PubMed] [Google Scholar]
  • 79.Protherics Inc . September 2010. Digifab (Digoxin Immune Fab [ovine]) Prescribing Information. Brentwood, TN. [Google Scholar]
  • 80.Smith T.W. Use of antibodies in the study of the mechanism of action of digitalis. Ann N Y Acad Sci. 1974;242:731–736. doi: 10.1111/j.1749-6632.1974.tb19134.x. [DOI] [PubMed] [Google Scholar]
  • 81.Watson J.F., Butler V.P. Biologic activity of digoxin-specific antisera. J Clin Investig. 1972;51:638–648. doi: 10.1172/JCI106853. ([PubMed]) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Butler V.P. Antibodies as specific antagonists of toxins, drugs, and hormones. Pharmacol Rev. 1982;34:109–114. [PubMed] [Google Scholar]
  • 83.Gardner J.D., Kiino D.R., Swartz T.J. Effects of digoxin-specific antibodies on accumulation and binding of digoxin by human erythrocytes. J Clin Investig. 1973;52:1820–1833. doi: 10.1172/JCI107364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sullivan J.B. Immunotherapy in the poisoned patient. Med Toxicol. 1986;1:47–60. doi: 10.1007/BF03259827. [DOI] [PubMed] [Google Scholar]
  • 85.Boucher B.A., Lalonde R.L. Digoxin-specific antibody fragments for the treatment of digoxin intoxication. Clin Pharm. 1986;5:826–827. [PubMed] [Google Scholar]
  • 86.Schmidt D.H., Butler V.P. Reversal of digoxin toxicity with specific antibodies. J Clin Investig. 1971;50:1738–1744. doi: 10.1172/JCI106663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Butler V.P., Schmidt D.H., Smith T.W. Effects of sheep digoxin-specific antibodies and their Fab fragments on digoxin pharmacokinetics in dogs. J Clin Investig. 1977;59:345–349. doi: 10.1172/JCI108647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Curd J.G., Smith T.W., Jaton J.C. The isolation of digoxin-specific antibody and its use in reversing the effects of digoxin. Proc Natl Acad Sci USA. 1971;68:2401–2406. doi: 10.1073/pnas.68.10.2401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Nisonoff A. Enzymatic digestion of rabbit gamma globulin and antibody and chromatography of digestion products. Methods Med Res. 1964;10:134–141. [PubMed] [Google Scholar]
  • 90.Lapostolle F., Borron S.W., Verdier C. Digoxin-specific Fab fragments as single first-line therapy in digitalis poisoning. Crit Care Med. 2008;36:3014–3018. doi: 10.1097/CCM.0b013e31818b341c. [DOI] [PubMed] [Google Scholar]
  • 91.Eyal D., Molczan K.A., Carroll L.S. Digoxin toxicity: pediatric survival after asystolic arrest. Clin Toxicol. 2005;43:51–54. doi: 10.1081/clt-45025. [DOI] [PubMed] [Google Scholar]
  • 92.Bateman D.N. Digoxin-specific antibody fragments: how much and when? Toxicol Rev. 2004;23:135–143. doi: 10.2165/00139709-200423030-00001. [DOI] [PubMed] [Google Scholar]
  • 93.Schaeffer T.H., Mlynarchek S.L., Stanford C.F. Treatment of chronically digoxin-poisoned patients with a newer digoxin immune fab--a retrospective study. J Am Osteopath Assoc. 2010;110:587–592. [PubMed] [Google Scholar]
  • 94.Ip D., Syed H., Cohen M. Digoxin specific antibody fragments (Digibind) in digoxin toxicity. BMJ. 2009;339:b2884. doi: 10.1136/bmj.b2884. [DOI] [PubMed] [Google Scholar]
  • 95.Flanagan R.J., Jones A.L. Fab antibody fragments: some applications in clinical toxicology. Drug Saf. 2004;27:1115–1133. doi: 10.2165/00002018-200427140-00004. [DOI] [PubMed] [Google Scholar]
  • 96.Ware J.A., Young J.B., Luchi R.J. Treatment of severe digoxin toxicity with digoxin-specific antibodies: a case report. Tex Med. 1983;79:57–59. [PubMed] [Google Scholar]
  • 97.Nicholls D.P., Murtagh J.G., Holt D.W. Use of amiodarone and digoxin specific Fab antibodies in digoxin overdosage. Br Heart J. 1985;53:462–464. doi: 10.1136/hrt.53.4.462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Centers for Disease Control and Prevention General recommendations on immunization: recommendations of the advisory committee on immunization practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2006;55(RR-15):1–47. [PubMed] [Google Scholar]
  • 99.American Academy of Pediatrics . 27th ed. American Academy of Pediatrics; Elk Grove Village, IL: 2006. 2006 Red Book: Report of the Committee on Infectious Diseases. [Google Scholar]
  • 100.Talecris . June 2007. HyperHEP B S/D (Hepatitis B Immune Globulin [human] Solvent/detergent Treated) Prescribing Information. [Google Scholar]
  • 101.Nabi . June 2003. Nabi-HB (Hepatitis B Immune Globulin [human] Solvent/detergent Treated and Filtered) Prescribing Information. Boca Raton, FL. [Google Scholar]
  • 102.Centers for Disease Control and Prevention A comprehensive immunization strategy to eliminate transmission of hepatitis B virus infection in the United States. Recommendations of the Advisory Committee on Immunization Practices (ACIP). Part I: immunization of infants, children, and adolescents. MMWR Recomm Rep (Morb Mortal Wkly Rep) 2005;54(RR-16):1–33. [PubMed] [Google Scholar]
  • 103.Centers for Disease Control and Prevention A comprehensive immunization strategy to eliminate transmission of hepatitis B virus infection in the United States. Recommendations of the Advisory Committee on Immunization Practices (ACIP). Part II: immunization in adults. MMWR Recomm Rep (Morb Mortal Wkly Rep) 2006;55(RR-16):1–33. [PubMed] [Google Scholar]
  • 104.Apotex . April 2007. HepaGam B (Hepatitis B Immune Globulin Intravenous [human]) Prescribing Information. Weston, FL. [Google Scholar]
  • 105.American Academy of Pediatrics . 27th ed. American Academy of Pediatrics; Elk Grove Village, IL: 2006. 2006 Red Book: Report of the Committee on Infectious Diseases. [Google Scholar]
  • 106.Centers for Disease Control and Prevention Recommended immunization schedules for persons 0 through 18 years–United States, 2009. MMWR Morb Mortal Wkly Rep. 2009;57 Q1-4. [Google Scholar]
  • 107.Centers for Disease Control Immunization Practices Advisory Committee (ACIP) Protection against viral hepatitis: recommendations of the immunization practices advisory committee (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 1990;39(RR-2):1–26. [PubMed] [Google Scholar]
  • 108.Centers for Disease Control and Prevention Sexually transmitted diseases treatment guidelines. MMWR Recomm Rep (Morb Mortal Wkly Rep) 2006;55(RR-11):1–94. [PubMed] [Google Scholar]
  • 109.Centers for Disease Control and Prevention Updated US Public Health Service guidelines for the management of occupational exposures to HBV, HCV, and HIV and recommendations for postexposure prophylaxis. MMWR Morb Mortal Wkly Rep. 2001;50(No. RR-11):1–51. [PubMed] [Google Scholar]
  • 110.Coffin C.S., Terrault N.A. Management of hepatitis B in liver transplant recipients. J Viral Hepat. 2007;14(Suppl1):37–44. doi: 10.1111/j.1365-2893.2007.00916.x. [DOI] [PubMed] [Google Scholar]
  • 111.Yilmaz N., Shiffman M.L., Stravitz R.T. Prophylaxis against recurrence of hepatitis B virus after liver transplantation: a retrospective analysis spanning 20 years. Liver Int. 2008;28:72–78. doi: 10.1111/j.1478-3231.2007.01611.x. [DOI] [PubMed] [Google Scholar]
  • 112.Gish R.G., McCashland T. Hepatitis B in liver transplant recipients. Liver Transplant. 2006;12:S54–S64. doi: 10.1002/lt.20950. [DOI] [PubMed] [Google Scholar]
  • 113.Anderson R.D., Chinnakotla S., Guo L. Intramuscular hepatitis B immunoglobulin (HBIG) and nucleosides for prevention of recurrent hepatitis B following liver transplantation: comparison with other HBIG regimens. Clin Transplant. 2007;21:510–517. doi: 10.1111/j.1399-0012.2007.00678.x. [DOI] [PubMed] [Google Scholar]
  • 114.Gane E.J., Angus P.W., Strasser S. Lamivudine plus low-dose hepatitis B immunoglobulin to prevent recurrent hepatitis B following liver transplantation. Gastroenterology. 2007;132:931–937. doi: 10.1053/j.gastro.2007.01.005. [DOI] [PubMed] [Google Scholar]
  • 115.Nath D.S., Kalis A., Nelson S. Hepatitis B prophylaxis post-liver transplant without maintenance hepatitis B immunoglobulin therapy. Clin Transplant. 2006;20:206–210. doi: 10.1111/j.1399-0012.2005.00467.x. [DOI] [PubMed] [Google Scholar]
  • 116.Eisenbach C., Sauer P., Mehrabi A. Prevention of hepatitis B virus recurrence after liver transplantation. Clin Transplant. 2006;20(Suppl):111–116. doi: 10.1111/j.1399-0012.2006.00609.x. [DOI] [PubMed] [Google Scholar]
  • 117.Zheng S., Chen Y., Liang T. Prevention of hepatitis B recurrence after liver transplantation using lamivudine or lamivudine combined with hepatitis B immunoglobulin prophylaxis. Liver Transplant. 2006;12:253–258. doi: 10.1002/lt.20701. [DOI] [PubMed] [Google Scholar]
  • 118.Cangene Corporation . December 2012. Varizig Varicella Zoster Immune Globulin (Human) Lyophilized Powder for Solution for Injection for Intramuscular Administration Only Prescribing Information. Winnipeg, Canada. [Google Scholar]
  • 119.Centers for Disease Control and Prevention Prevention of varicella: recommendations of the advisory committee on immunization practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2007;56(RR-4):1–40. [PubMed] [Google Scholar]
  • 120.National Center for Immunization and Respiratory Diseases General recommendations on immunization --- recommendations of the advisory committee on immunization practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2011;60:1–64. [PubMed] [Google Scholar]
  • 121.American Academy of Pediatrics . 28th ed. American Academy of Pediatrics; Elk Grove Village, IL: 2009. Red Book: 2009 Report of the Committee on Infectious Diseases. [Google Scholar]
  • 122.Centers for Disease Control and Prevention (CDC) FDA approval of an extended period for administering VariZIG for postexposure prophylaxis of varicella. MMWR Morb Mortal Wkly Rep. 2012;61:212. [PubMed] [Google Scholar]
  • 123.Solstice Neurosciences . May 2010. Myobloc (rimabotulinumtoxinB) Injection Prescribing Information. South San Francisco, CA. [Google Scholar]
  • 124.Brashear A., Lew M.F., Dykstra D.D. Safety and efficacy of NeuroBloc (botulinum toxin type B) in type A-responsive cervical dystonia. Neurology. 1999;53:1439–1446. doi: 10.1212/wnl.53.7.1439. [DOI] [PubMed] [Google Scholar]
  • 125.Terranova W., Breman J.G., Locey R.P. Botulism type B: epidemiologic aspects of an extensive outbreak. Am J Epidemiol. 1978;108:150–156. doi: 10.1093/oxfordjournals.aje.a112599. [DOI] [PubMed] [Google Scholar]
  • 126.Cheng C.M., Chen J.S., Patel R.P. Unlabeled uses of botulinum toxins: a review, part 1. Am J Health Syst Pharm. 2006;63:145–152. doi: 10.2146/ajhp050137. [DOI] [PubMed] [Google Scholar]
  • 127.Allergan . September 2013. Botox (onabotulinumtoxinA) for Injection Prescribing Information. Irvine, CA. [Google Scholar]
  • 128.Bell M.S., Vermeulen L.C., Sperling K.B. Pharmacotherapy with botulinum toxin: harnessing nature's most potent neurotoxin. Pharmacotherapy. 2000;20:1079–1091. doi: 10.1592/phco.20.13.1079.35040. [DOI] [PubMed] [Google Scholar]
  • 129.Tsui J.K. Botulinum toxin as a therapeutic agent. Pharmacol Ther. 1996;72:13–24. doi: 10.1016/s0163-7258(96)00091-5. [DOI] [PubMed] [Google Scholar]
  • 130.Moore A.P. Botulinum toxin A (BoNT-A) for spasticity in adults. What is the evidence? Eur J Neurol. 2002;9(Suppl 1):42–47. doi: 10.1046/j.1468-1331.2002.0090s1042.x. [DOI] [PubMed] [Google Scholar]
  • 131.Corry I.S., Cosgrove A.P., Duffy C.M. Botulinum toxin A compared with stretching casts in the treatment of spastic equinus: a randomised prospective trial. J Pediatr Orthop. 1998;18:304–311. [PubMed] [Google Scholar]
  • 132.Figgit D.P., Noble S. Botulinum toxin B: a review of its therapeutic potential in the management of cervical dystonia. Drugs. 2002;62:705–722. doi: 10.2165/00003495-200262040-00011. [DOI] [PubMed] [Google Scholar]
  • 133.Lew M.F., Adornato B.T., Duane D.D. Botulinum toxin type B: a double-blind, placebo-controlled, safety and efficacy study in cervical dystonia. Neurology. 1997;49:701–707. doi: 10.1212/wnl.49.3.701. [DOI] [PubMed] [Google Scholar]
  • 134.Anon Botulinum toxin for cervical dystonia. Med Lett Drugs Ther. 2001;43:63–64. [PubMed] [Google Scholar]
  • 135.California Department of Public Health . January 2012. BabyBIG (Botulism Immune Globulin Intravenous [human]) Prescribing Information. Richmond, CA. [Google Scholar]
  • 136.Arnon S.S. Creation and development of the public service orphan drug Human Botulism Immune Globulin. Pediatrics. 2007;119:785–789. doi: 10.1542/peds.2006-0646. ([PubMed]) [DOI] [PubMed] [Google Scholar]
  • 137.Infant Botulism Treatment and Prevention Program. Division of Communicable Disease Control, California Department of Health Services. From IBTPP website. Accessed 2012 Mar 26. [Web].
  • 138.Underwood K., Rubin S., Deakers T. Infant botulism: a 30-year experience spanning the introduction of botulism immune globulin intravenous in the intensive care unit at Childrens Hospital Los Angeles. Pediatrics. 2007;120:e1380–e1385. doi: 10.1542/peds.2006-3276. ([PubMed]) [DOI] [PubMed] [Google Scholar]
  • 139.Arnon S.S. Infant botulism. In: Feigin R.D., Cherry J.D., Demmler-Harrison G.J., editors. Feigin: Feigin and Cherry's Textbook of Pediatric Infectious Diseases. 6th ed. Saunders Elsevier; Philadelphia, PA: 2009. [Google Scholar]
  • 140.Arnon S.S., Schechter R., Maslanka S.E. Human botulism immune globulin for the treatment of infant botulism. N Engl J Med. 2006;354:462–471. doi: 10.1056/NEJMoa051926. [DOI] [PubMed] [Google Scholar]
  • 141.Talecris Biotherapeutics, Inc . Research Triangle Park; NC: March 2008. HyperRAB S/D (Rabies Immune Globulin [human] Solvent/detergent Treated) Prescribing Information. [Google Scholar]
  • 142.Sanofi Pasteur . December 2005. Imogam Rabies-HT (Rabies Immune Globulin [human] USP, Heat Treated) Prescribing Information. Swiftwater, PA. [Google Scholar]
  • 143.Centers for Disease Control and Prevention Human rabies prevention—United States, 2008. Recommendations of the advisory committee on immunization practices. MMWR Recomm Rep (Morb Mortal Wkly Rep) 2008;57(RR-3):1–27. [PubMed] [Google Scholar]
  • 144.Centers for Disease Control and Prevention Use of a reduced (4-dose) vaccine schedule for postexposure prophylaxis to prevent human rabies. Recommendations of the Advisory Committee on Immunization Practices. MMWR Recomm Rep (Morb Mortal Wkly Rep) 2010;59(RR-2):1–9. [PubMed] [Google Scholar]
  • 145.Cangene bioPharma . December 2010. WinRho SDF (Rho [D] Immune Globulin Intravenous [human]) Prescribing Information. Baltimore, MD. [Google Scholar]
  • 146.Grifols Therapeutics . Research Triangle Park; NC: September 2012. HyperRHO S/D Mini-Dose (Rho [D] Immune Globulin [human]) Prescribing Information. [Google Scholar]
  • 147.Ortho-Clinical Diagnostics . Raritan; NJ: November 2012. Rho-GAM (Rho [D] Immune Globulin [human]) Ultra-filtered PLUS and MICRhoGAM (Rho [D] Immune Globulin [human]) Ultra-filtered PLUS Prescribing Information. [Google Scholar]
  • 148.The United States pharmacopeia . 18th ed. The United States Pharmacopeial Convention, Inc; Rockville, MD: 1995. 23rd Rev, and the National Formulary; p. 350. [Google Scholar]
  • 149.CSL Behring . Kankakee; IL: October 2012. Rhophylac (Rho [D] Immune Globulin Intravenous [human]) Prescribing Information. [Google Scholar]
  • 150.Kumar S. Universal RDH genotyping in fetuses. BMJ. 2008;336:783. doi: 10.1136/bmj.39533.358252.BE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Bussel J.B., Graziano J.N., Kimberly R.P. Intravenous anti-D treatment of immune thrombocytopenic purpura: analysis of efficacy, toxicity, and mechanism of effect. Blood. 1991;77:1884–1893. [PubMed] [Google Scholar]
  • 152.Becker T., Küenzlen E., Salama A. Treatment of childhood idiopathic thrombocytopenic purpura with Rhesus antibodies (anti-D) Eur J Pediatr. 1986;145:166–169. doi: 10.1007/BF00446055. [DOI] [PubMed] [Google Scholar]
  • 153.Andrew M., Blanchette V.S., Adams M. A multicenter study of the treatment of childhood chronic idiopathic thrombocytopenic purpura with anti-D. J Pediatr. 1992;120:522–527. doi: 10.1016/s0022-3476(10)80001-0. [DOI] [PubMed] [Google Scholar]
  • 154.Scaradavou A., Woo B., Woloski B.M.R. Intravenous anti-D treatment of immune thrombocytopenic 20. Ballow M. Mechanisms of action of intravenous immunoglobulin therapy and potential use in autoimmune connective tissue diseases. Cancer. 1991;68:1430–1436. doi: 10.1002/1097-0142(19910915)68:6+<1430::aid-cncr2820681405>3.0.co;2-4. [DOI] [PubMed] [Google Scholar]
  • 155.Ballow M. Mechanisms of action of intravenous immunoglobulin therapy and potential use in autoimmune connective tissue diseases. Cancer. 1991;68:1430–1436. doi: 10.1002/1097-0142(19910915)68:6+<1430::aid-cncr2820681405>3.0.co;2-4. [DOI] [PubMed] [Google Scholar]
  • 156.Kniker W.T. Immunosuppressive agents, γ-globulin, immunomodulation, immunization, and aphresis. J Allergy Clin Immunol. 1989;84:1104–1107. doi: 10.1016/0091-6749(89)90163-2. [DOI] [PubMed] [Google Scholar]
  • 157.Blanchette V., Imbach P., Andrew M. Randomised trial of intravenous immunoglobulin G, intravenous anti-D, and oral prednisone in childhood acute immune thrombocytopenic purpura. Lancet. 1994;344:703–707. doi: 10.1016/s0140-6736(94)92205-5. [DOI] [PubMed] [Google Scholar]
  • 158.Berchtold P., McMillan R. Therapy of chronic idiopathic thrombocytopenic purpura in adults. Blood. 1989;74:2309–2317. [PubMed] [Google Scholar]
  • 159.Rodeghiero F., Schiavotto C., Castaman G. A follow-up study of 49 adult patients with idiopathic thrombocytopenic purpura treated with high-dose immunoglobulins and anti-D immunoglobulins. Haematologica. 1992;77:248–252. [PubMed] [Google Scholar]
  • 160.Stasi R., Stipa E., Masi M. Long-term observation of 208 adults with chronic idiopathic thrombocytopenic purpura. Am J Med. 1995;98:436–442. doi: 10.1016/s0002-9343(99)80342-8. [DOI] [PubMed] [Google Scholar]
  • 161.Landonio G., Galli M., Nosari A. HIV-related severe thrombocytopenia in intravenous drug-users: prevalence, response to therapy in a medium-term follow-up, and pathogenetic evaluation. AIDS. 1990;4:29–34. [PubMed] [Google Scholar]
  • 162.Hoffman D.M., Caruso R.F., Mirando T. Human immunodeficiency virus-associated thrombocytopenia. Dicp Ann Pharmacother. 1989:157–160. doi: 10.1177/106002808902300212. [DOI] [PubMed] [Google Scholar]
  • 163.Food and Drug Administration. FDA Application: Search Orphan Drug Designations and Approvals. Silver Spring, MD. From FDA website (http://www.accessdata.fda.gov/scripts/opdlisting/oopd/index.cfm). Accessed 2013 Jul 2.
  • 164.Oksenhendler E., Bierling P., Brossard Y. Anti-RH immunoglobulin therapy for human immunodeficiency virus-related immune thrombocytopenic purpura. Blood. 1988;71:1499–1502. [PubMed] [Google Scholar]
  • 165.Okwundu C.I., Afolabi B.B. Intramuscular versus intravenous anti-D for preventing Rhesus alloimmunization during pregnancy. Cochrane Database Syst Rev. 2013;1:CD007885. doi: 10.1002/14651858.CD007885.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.CSL Behring . Kankakee; IL: November 2016. Carimune NF, Nanofiltered (Immune Globulin Intravenous [human] Lyophilized for Solution) Prescribing Information. [Google Scholar]
  • 167.Baxalta US Inc . September 2016. Gammagard S/D (Immune Globulin Intravenous [human] IgA Less than or Equal to 2.2 mcg/mL in a 5% Solution) Prescribing Information. Westlake Village, CA. [Google Scholar]
  • 168.Grifols USA . Research Triangle Park; NC: June 2017. GamaSTAN S/D (Immune Globulin IM [human]) Prescribing Information. [Google Scholar]
  • 169.Octapharma USA . August 2015. Octagam (Immune Globulin Intravenous [human] 5% Liquid) Prescribing Information. Hoboken, NJ. [Google Scholar]
  • 170.Grifols Therapeutics Inc . Research Triangle Park; NC: September 2016. Gamunex-C (Immune Globulin Intravenous [human] 10% Caprylate/chromatography Purified) Prescribing Information. [Google Scholar]
  • 171.Baxalta US Inc . September 2016. Gammagard S/D (Immune Globulin Intravenous [human] IgA Less than 1 mcg/mL in a 5% Solution) Prescribing Information. Westlake Village, CA. [Google Scholar]
  • 172.CSL Behring . October 2016. Privigen (Immune Globulin Intravenous [human] 10% Liquid) Prescribing Information. Kankakee, IL. [Google Scholar]
  • 173.American Academy of Pediatrics . 30th ed. American Academy of Pediatrics; Elk Grove Village, IL: 2015. Red Book: 2015 Report of the Committee on Infectious Diseases. [Google Scholar]
  • 174.NIH Consensus Development Conference. Intravenous immunoglobulin: prevention and treatment of disease. J Am Med Assoc. 1990;264:3189–3193. [PubMed] [Google Scholar]
  • 175.Hughes R.A., Donofrio P., Bril V. Intravenous immune globulin (10% caprylate-chromatography purified) for the treatment of chronic inflammatory demyelinating polyradiculoneuropathy (ICE study): a randomised placebo-controlled trial. Lancet Neurol. 2008;7:136–144. doi: 10.1016/S1474-4422(07)70329-0. [DOI] [PubMed] [Google Scholar]
  • 176.CSL Behring . October 2016. Hizentra (Immune Globulin Subcutaneous [human] 20% Liquid) Prescribing Information. Kankakee, IL. [Google Scholar]
  • 177.Bio Products Laboratory (distributed by BPL Inc) September 2015. Gammaplex (Immune Globulin Intravenous [human] 5% Liquid for Intravenous Use) Prescribing Information. Hertfordshire, UK. [Google Scholar]
  • 178.Biotest Pharmaceuticals . April 2014. Bivigam (Immune Globulin Intravenous [human] 10% Liquid) Prescribing Information. Boca Raton, FL. [Google Scholar]
  • 179.Grifols USA . January 2016. Flebogamma 10% DIF (Immune Globulin Intravenous [human] Solution for Intravenous Administration) Prescribing Information. Los Angeles, CA. [Google Scholar]
  • 180.Octapharma USA . November 2015. Octagam (Immune Globulin Intravenous [human] 10% Liquid) Prescribing Information. Hoboken, NJ. [Google Scholar]
  • 181.Baxalta US Inc . April 2016. Hyqvia (Immune Globulin [human] 10% with Recombinant Human Hyaluronidase Solution for Subcutaneous Administration) Prescribing Information. Westlake Village, CA. [Google Scholar]
  • 182.Baxalta US Inc . September 2016. Cuvitru (Immune Globulin Subcutaneous [human] 20% Solution) Prescribing Information. Westlake Village, CA. [Google Scholar]
  • 183.Kedrion Biopharma . September 2016. Gammaked (Immune Globulin Intravenous [human] 10% Caprylate/chromatography Purified) Prescribing Information. Fort Lee, NJ. [Google Scholar]
  • 184.Advisory Committee on Immunization Practices (ACIP), Fiore A.E., Wasley A. Prevention of hepatitis A through active or passive immunization: recommendations of the advisory committee on immunization practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2006;55(RR-7):1–23. [PubMed] [Google Scholar]
  • 185.Centers for Disease Control and Prevention Update: prevention of hepatitis A after exposure to hepatitis A virus and in international travelers. Updated recommendations of the advisory committee on immunization practices (ACIP) MMWR Morb Mortal Wkly Rep. 2007;56:1080–1084. [PubMed] [Google Scholar]
  • 186.Nelson N.P. Updated dosing instructions for immune globulin (human) GamaSTAN S/D for hepatitis a virus prophylaxis. MMWR Morb Mortal Wkly Rep. 2017;66:959–960. doi: 10.15585/mmwr.mm6636a5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Centers for Disease Control and Prevention . US Department of Health and Human Services; Atlanta, GA: 2018. CDC Health Information for International Travel. (Updates may be available at: CDC website) [Google Scholar]
  • 188.McLean H.Q., Fiebelkorn A.P., Temte J.L. Prevention of measles, rubella, congenital rubella syndrome, and mumps, 2013: summary recommendations of the Advisory Committee on Immunization Practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2013;62(RR-04):1–34. [PubMed] [Google Scholar]
  • 189.Gardulf A., Nocolay U., Asensio O. Rapid subcutaneous IgG replacement therapy is effective and safe in children and adults with primary immunodeficiencies-a prospective, multi-national study. J Clin Immunol. 2006;26:177–185. doi: 10.1007/s10875-006-9002-x. [DOI] [PubMed] [Google Scholar]
  • 190.Nicolay U., Kiessling P., Berger M. Health-related quality of life and treatment satisfaction in north american patients with primary immunedeficiency diseases receiving subcutaneous IgG self-infusions at home. J Clin Immunol. 2006;26:65–72. doi: 10.1007/s10875-006-8905-x. [DOI] [PubMed] [Google Scholar]
  • 191.Gaspar J., Berritsen B., Jones A. Immunoglobulin replacement treatment by rapid subcutaneous infusion. BMJ. 1998;79:48–51. doi: 10.1136/adc.79.1.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Grifols USA . April 2015. Flebogamma 5% DIF (Immune Globulin Intravenous [human] Solution for Intravenous Administration) Prescribing Information. Los Angeles, CA. [Google Scholar]
  • 193.Kurtsberg J., Friedman H.S., Chaffee S. Efficacy of intravenous gamma globulin in autoimmune-mediated pediatric blood dyscrasias. Am J Med. 1987;83(Suppl 4A):4–9. doi: 10.1016/0002-9343(87)90544-4. [DOI] [PubMed] [Google Scholar]
  • 194.Lusher J.M., Warrier I. Use of intravenous gamma globulin in children and adolescents with idiopathic thrombocytopenic purpura and other immune thrombocytopenias. Am J Med. 1987;83(Suppl 4A):10–16. doi: 10.1016/0002-9343(87)90545-6. [DOI] [PubMed] [Google Scholar]
  • 195.Berkman S.A., Lee M.L., Gale R.P. Clinical uses of intravenous immunoglobulins. Ann Intern Med. 1990;112:278–292. doi: 10.7326/0003-4819-112-4-278. [DOI] [PubMed] [Google Scholar]
  • 196.Cooperative Group for the Study of Immunoglobulin in Chronic Lymphocytic Leukemia Intravenous immunoglobulin for the prevention of infection in chronic lymphocytic leukemia. N Engl J Med. 1988;319:902–907. doi: 10.1056/NEJM198810063191403. [DOI] [PubMed] [Google Scholar]
  • 197.Knapp M.J., Colburn P.A. Clinical uses of intravenous immune globulin. Clin Pharm. 1990;9:509–529. [PubMed] [Google Scholar]
  • 198.Newburger J.W., Takahashi M., Burns J.C. The treatment of Kawasaki syndrome with intravenous gamma globulin. N Engl J Med. 1986;315:341–347. doi: 10.1056/NEJM198608073150601. [DOI] [PubMed] [Google Scholar]
  • 199.Nagashima M., Matsushima M., Matsuoka H. High-dose gammaglobulin therapy for Kawasaki disease. J Pediatr. 1987;110:710–712. doi: 10.1016/s0022-3476(87)80007-0. [DOI] [PubMed] [Google Scholar]
  • 200.Newburger J.W., Takahashi M., Gerber M.A. Diagnosis, treatment, and long-term management of Kawasaki disease: a statement for health professionals from the committee on rheumatic fever, endocarditis and Kawasaki disease, council on cardiovascular disease in the young, American heart association. Circulation. 2004;110:2747–2771. doi: 10.1161/01.CIR.0000145143.19711.78. [DOI] [PubMed] [Google Scholar]
  • 201.Singh S., Tank N.K., Dwiwedi P., Charan J. Monoclonal antibodies: a review. Curr Clin Pharmacol. 2018;13:85–99. doi: 10.2174/1574884712666170809124728. PMID:28799485. [DOI] [PubMed] [Google Scholar]
  • 202.HIGHLIGHTS OF PRESCRIBING INFORMATION: RITUXAN HYCELA (rituximab and hyaluronidase human) injection, for subcutaneous use Initial U.S. Approval: 2017. Revised 6/2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761064s000lbl.pdf.
  • 203.Salles G., Barrett M., Robin Foà R. Rituximab in B-cell hematologic malignancies: a review of 20 Years of clinical experience. Adv Ther. 2017;34:2232–2273. doi: 10.1007/s12325-017-0612-x. PMID:28983798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Qi J., Chen S., Chiorazzi N., Rader C. An IgG1-like bispecific antibody targeting CD52 and CD20 for the treatment of B-cell malignancies. Methods. 2019;154:70–76. doi: 10.1016/j.ymeth.2018.08.008. https://DOI.org/10.1016/j.ymeth.2018.08.008 Available online 24 August 2018 at. [DOI] [PubMed] [Google Scholar]
  • 205.Zhang B. Mini-review: ofatumumab. mAbs. 2009;1(4):326–331. doi: 10.4161/mabs.1.4.8895. http://www.landesbioscience.com/journals/mabs/article/8895 Previously published online as a mAbs E-publication: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.McWilliams E.M., Meleb J.M., Cheney C. Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia. OncoImmunology. 2016;5(10):e1226720. doi: 10.1080/2162402X.2016.1226720. (9 pages) Accepted 16 August 2016. Available at: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Robak T., Hellmann A., Kloczko J. Randomized phase 2 study of otlertuzumab and bendamustine versus bendamustine in patients with relapsed chronic lymphocytic leukaemia. Br J Haematol. 2017;176:618–628. doi: 10.1111/bjh.14464. PMID: 24843434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Segal N.H., Logan T.F., Hodi S. Results from an integrated safety analysis of urelumab, an agonist anti-CD137 monoclonal antibody. Clin Cancer Res. 2017;23(8):1929–1936. doi: 10.1158/1078-0432.CCR-16-1272. http://clincancerres.aacrjournals.org/content/23/8/1929 Published OnlineFirst October 18, 2016. Available at: [DOI] [PubMed] [Google Scholar]
  • 209.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; Urelumab (CD137 mAb) with rituximab for relapsed, refractory or high-risk untreated chronic lymphocytic leukemia (CLL) patients. Available at: https://clinicaltrials.gov/ct2/show/NCT02420938?term=urelumab&rank=1.
  • 210.Kashyap M.K., Kumar D., Jones H. Ulocuplumab (BMS-936564/MDX1338): a fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway. Oncotarget. January 19, 2016;7(3):2809–2822. doi: 10.18632/oncotarget.6465. Published online 2015 Dec 4. PMID:26646452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.First in Human Study to Determine the Safety, Tolerability and Preliminary Efficacy of an Anti-CXCR4 Antibody in Subjects With Acute Myelogenous Leukemia and Selected B-cell Cancers. Available at: https://clinicaltrials.gov/ct2/show/NCT01120457?term=ulocuplumab&rank=5.
  • 212.Lin T.S., Stock W., Xu H. A phase I/II dose escalation study of apolizumab (Hu1D10) using a stepped-up dosing schedule in patients with chronic lymphocytic leukemia and acute leukemia. Leuk Lymphoma. 2009;50(12):1958–1963. doi: 10.3109/10428190903186486. PMID: 19860603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.de Vos S., Forero-Torres A., Ansell S.M. A phase II study of dacetuzumab (SGN-40) in patients with relapsed diffuse large B-cell lymphoma (DLBCL) and correlative analyses of patient-specific factors. J Hematol Oncol. 2014;7:44. doi: 10.1186/1756-8722-7-44. PMID:24919462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Fayad L., Ansell S.M., Advani R. Dacetuzumab plus rituximab, ifosfamide, carboplatin and etoposide as salvage therapy for patients with diffuse large B-cell lymphoma relapsing after rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone: a randomized, double-blind, placebo-controlled phase 2b trial. Leuk Lymphoma. 2015;56(9):2569–2578. doi: 10.3109/10428194.2015.1007504. PMID:25651427. [DOI] [PubMed] [Google Scholar]
  • 215.Awan F.T., Hillmen P., Hellmann A. A randomized, open-label, multicentre, phase 2/3 study to evaluate the safety and efficacy of lumiliximab in combination with fludarabine, cyclophosphamide and rituximab versus fludarabine, cyclophosphamide and rituximab alone in subjects with relapsed chronic lymphocytic leukaemia. Br J Haematol. 2014;167:466–477. doi: 10.1111/bjh.13061. PMID:25130401. [DOI] [PubMed] [Google Scholar]
  • 216.Kovtun Y., Jones G.E., Adams S. A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells. Blood Adv. April 24, 2018;2(8):848–858. doi: 10.1182/bloodadvances.2018017517. PMID:29661755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Aigner M., Feulner J., Schaffer S. T lymphocytes can be effectively recruited for ex vivo and in vivo lysis of AML blasts by a novel CD33/CD3-bispecific BiTE antibody construct. Leukemia. April 2013;27(5):1107–1115. doi: 10.1038/leu.2012.341. Epub 2012 Nov 26. PMID:23178753. [DOI] [PubMed] [Google Scholar]
  • 218.Xie L.H., Biondo M., Busfield S.J. CD123 target validation and preclinical evaluation of ADCC activity of anti-CD123 antibody CSL362 in combination with NKs from AML patients in remission. Blood Canc J. 2017;7:e567. doi: 10.1038/bcj.2017.52. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5520399/ published online 2 June 2017. PMID:28574487. Available at: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; Study of Biomarker-Based Treatment of Acute Myeloid Leukemia. Available at: https://clinicaltrials.gov/ct2/show/NCT03013998?term=samalizumab&rank=3.
  • 220.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; Ficlatuzumab With High Dose Cytarabine in Relapsed and Refractory AML available at: https://clinicaltrials.gov/ct2/show/NCT02109627?term=Ficlatuzumab&rank=3.
  • 221.Egan P.C., Reagan J.L. The return of gemtuzumab ozogamicin: a humanized anti-CD33 monoclonal antibody–drug conjugate for the treatment of newly diagnosed acute myeloid leukemia. OncoTargets Ther. 2018;11:8265–8272. doi: 10.2147/OTT.S150807. eCollection 2018. PMID:30538495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Feldman E.J., Brandwein J., Stone R. Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, Lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia. J Clin Oncol. June 20, 2005;23(18):4110–4116. doi: 10.1200/JCO.2005.09.133. PMID: 15961759. [DOI] [PubMed] [Google Scholar]
  • 223.Sekeres M.A., Lancet J.E., Wood B.L. Randomized, phase IIb study of low-dose cytarabine and lintuzumab versus low-dose cytarabine and placebo in older adults with untreated acute myeloid leukemia. Haematologica. January 2013;98(1):119–128. doi: 10.3324/haematol.2012.066613. Epub 2012 Jul 16. PMID:22801961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.de Weers M., Tai Y.T., van der Veer M.S. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011;186(3):1840–1848. doi: 10.4049/jimmunol.1003032. http://www.jimmunol.org/content/186/3/1840 Epub 2010 Dec 27. PMID:21187443. Available at: [DOI] [PubMed] [Google Scholar]
  • 225.HIGHLIGHTS OF PRESCRIBING INFORMATION DARZALEX (daratumumab) injection. Revised 11/2016 Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/761036s004lbl.pdf.
  • 226.Shah J.J., Feng L., Thomas S.K. Siltuximab (CNTO 328) with lenalidomide, bortezomib and dexamethasone in newly-diagnosed, previously untreated multiple myeloma: an open-label phase I trial. Blood Canc J. 2016;6:e396. doi: 10.1038/bcj.2016.4. published online 12 February 2016. PMID:26871714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.CENTER FOR DRUG EVALUATION AND RESEARCH; Approval Package for: APPLICATION NUMBER: 125496Orig1s000 Trade Name: Sylvant, Generic Name: siltuximab, Sponsor: Janssen Biotech, Inc, Approval Date: April 23, 2014. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/125496Orig1s000Approv.pdf.
  • 228.Le Jeune C., Thomas X. Potential for bispecific T-cell engagers: role of blinatumomab in acute lymphoblastic leukemia. Drug Des Dev Ther. February 18, 2016;10:757–765. doi: 10.2147/DDDT.S83848. eCollection 2016. PMID:26937176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Kantarjian H., Stein A., Gökbuget N. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. March 02, 2017;376(9):836–847. doi: 10.1056/NEJMoa1609783. PMID: 28249141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.HIGHLIGHTS OF PRESCRIBING INFORMATION; BLINCYTO (blinatumomab) for injection, for intravenous use; Initial U.S. Approval: 2014 Revised 7/2017 https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125557s008lbl.pdf.
  • 231.Scott L.J. Brentuximab vedotin: a review in CD30-positive Hodgkin lymphoma. Drugs. 2017;77:435–445. doi: 10.1007/s40265-017-0705-5. Published online: 11 February 2017. PMID:28190142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Flynn M.J., Zammarchi F., Tyrer P.C. ADCT-301, a pyrrolobenzodiazepine (PBD) dimer–containing antibody–drug conjugate (ADC) Targeting CD25-expressing hematological malignancies. Mol Cancer Ther. November 2016;15(11):2709–2721. doi: 10.1158/1535-7163.MCT-16-0233. Epub 2016 Aug 17. PMID:27535974. [DOI] [PubMed] [Google Scholar]
  • 233.Study of ADCT-301 in Patients With Relapsed or Refractory Hodgkin and Non-Hodgkin Lymphoma. Available at: https://clinicaltrials.gov/ct2/show/NCT02432235?term=Camidanlumab+tesirine&rank=3.
  • 234.Ansell S.M., Horwitz S.M., Engert A. Phase I/II study of an anti-CD30 monoclonal antibody (MDX-060) in Hodgkin's Lymphoma and anaplastic large-cell lymphoma. J Clin Oncol. 2007;25:2764–2769. doi: 10.1200/JCO.2006.07.8972. PMID:17515574. [DOI] [PubMed] [Google Scholar]
  • 235.Fanale M., Assouline S., Kuruvilla J. Phase IA/II, multicentre, open-label study of the CD40 antagonistic monoclonal antibody lucatumumab in adult patients with advanced non-Hodgkin or Hodgkin lymphoma. Br J Haematol. January 2014;164(2):258–265. doi: 10.1111/bjh.12630. PMID: 24219359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Pro B., Advani R., Brice P. Five-year results of brentuximab vedotin in patients with relapsed or refractory systemic anaplastic large cell lymphoma. Blood. July 26, 2018;132(4):458–459. doi: 10.1182/blood-2017-05-780049. PMID:30049735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Schmid P., Adams S., Rugo H.S. Atezolizumab and Nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108–2121. doi: 10.1056/NEJMoa1809615. https://www.nejm.org/doi/full/10.1056/NEJMoa1809615 Available at: [DOI] [PubMed] [Google Scholar]
  • 238.Adams S., Diamond J.R., Hamilton E. Atezolizumab Plus nab-Paclitaxel in the treatment of metastatic triple-negative breast cancer with 2-year survival follow-up a phase 1b clinical trial. JAMA Oncol. October 19, 2018 doi: 10.1001/jamaoncol.2018.5152. [Epub ahead of print]. PMID:30347025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Cremolini C., Loupakis F., Antoniotti C. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol. 2015;16:1306–1315. doi: 10.1016/S1470-2045(15)00122-9. Available at: [DOI] [PubMed] [Google Scholar]
  • 240.HIGHLIGHTS OF PRESCRIBING INFORMATION; AVASTIN (bevacizumab) injection, for intravenous use; Initial U.S. Approval: 2004 Revised 12/2017; Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125085s319lbl.pdf.
  • 241.Inman B.A., Longo T.A., Ramalingam S. Atezolizumab: a PD-L1–blocking antibody for bladder cancer. Clin Cancer Res. 2017;23:1886–1890. doi: 10.1158/1078-0432.CCR-16-1417. Published OnlineFirst November 30, 2016. PMID:27903674. [DOI] [PubMed] [Google Scholar]
  • 242.Petrylak D.P., Powles T., Bellmunt J. Atezolizumab (MPDL3280A) monotherapy for patients with metastatic urothelial cancer long-term outcomes from a phase 1 study. JAMA Oncol. April 1, 2018;4(4):537–544. doi: 10.1001/jamaoncol.2017.5440. PMID:29423515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Horn L., Spigel D.R., Vokes E.E. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057) J Clin Oncol. 2017;35:3924–3933. doi: 10.1200/JCO.2017.74.3062. Available at: DOI: [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Hodi F.S., Chiarion-Sileni V., Gonzalez R. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018;19(11):1480–1492. doi: 10.1016/S1470-2045(18)30700-9. Epub 2018 Oct 22. PMID:30361170. [DOI] [PubMed] [Google Scholar]
  • 245.Cella D., Grünwald V., Escudier B. Patient-reported outcomes of patients with advanced renal cell carcinoma treated with nivolumab plus ipilimumab versus sunitinib (CheckMate 214): a randomised, phase 3 trial. Lancet Oncol. February 2019;20(2):297–310. doi: 10.1016/S1470-2045(18)30778-2. Epub 2019 Jan 15. PMID:30658932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Coleman R.L., Brady M.F., Herzog T.J. Bevacizumab and paclitaxel–carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. June 2017;18(6):779–791. doi: 10.1016/S1470-2045(17)30279-6. Epub 2017 Apr 21. PMID:28438473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Kaufman H.L., Russell J., Hamid O. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol. October 2016;17(10):1374–1385. doi: 10.1016/S1470-2045(16)30364-3. Epub 2016 Sep 1. PMID:27592805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.HIGHLIGHTS OF PRESCRIBING INFORMATION; BAVENCIO (avelumab) injection, for intravenous use Initial U.S. Approval: 2017. Revised 3/2017; Available at:: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761049s000lbl.pdf.
  • 249.NIH: National Library of Medicine at Clinicaltrials.gov for 175 Studies found for: Avelumab; Also searched for MSB0010718C and Bavencio. See search details available at: https://clinicaltrials.gov/ct2/results?cond=&term=Avelumab&cntry=&state=&city=&dist=.
  • 250.Ploessl C., Pan A., Maples K.T. Dinutuximab: an anti-GD2 monoclonal antibody for high-risk neuroblastoma. Ann Pharmacother. May 2016;50(5):416–422. doi: 10.1177/1060028016632013. Epub 2016 Feb 25. PMID:26917818. [DOI] [PubMed] [Google Scholar]
  • 251.Dhillon S. Dinutuximab: first global approval. Drugs. May 2015;75(8):923–927. doi: 10.1007/s40265-015-0399-5. PMID:25940913. [DOI] [PubMed] [Google Scholar]
  • 252.Diaz R.J., Ali S., Qadir M.G. The role of bevacizumab in the treatment of glioblastoma. J Neuro Oncol. July 2017;133(3):455–467. doi: 10.1007/s11060-017-2477-x. Epub 2017 May 19. PMID:28527008. [DOI] [PubMed] [Google Scholar]
  • 253.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; Anti-LAG-3 Alone & in Combination w/Nivolumab Treating Patients w/Recurrent GBM (Anti-CD137 Arm Closed 10/16/18) Available at: https://www.clinicaltrials.gov/ct2/show/NCT02658981?term=BMS-986016&rank=7.
  • 254.Lee S.J., Lee S.Y., Lee W.S. Phase I trial and pharmacokinetic study of tanibirumab, a fully human monoclonal antibody to vascular endothelial growth factor receptor 2, in patients with refractory solid tumors. Investig New Drugs. December 2017;35(6):782–790. doi: 10.1007/s10637-017-0463-y. Epub 2017 Apr 8. PMID:28391576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; Trial to Evaluate the Safety of TTAC-0001(Tanibirumab) in Recurrent Glioblastoma. Available at: https://www.clinicaltrials.gov/ct2/show/NCT03033524?term=tanibirumab&rank=2.
  • 256.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; TTAC-0001 Phase II Trial With Recurrent Glioblastoma Progressed on Bevacizumab. Available at: https://www.clinicaltrials.gov/ct2/show/NCT03856099?term=ttac-0001&rank=1.
  • 257.Seimetz D., Lindhofer H., Bokemeyer C. Development and approval of the trifunctional antibody catumaxomab (anti-EpCAM anti-CD3) as a targeted cancer immunotherapy. Cancer Treat Rev. October 2010;36(6):458–467. doi: 10.1016/j.ctrv.2010.03.001. Epub 2010 Mar 27. Review. PMID:20347527. [DOI] [PubMed] [Google Scholar]
  • 258.Seimetz D. Novel monoclonal antibodies for cancer treatment: the trifunctional antibody catumaxomab (Removab) J Cancer. 2011;2:309–316. doi: 10.7150/jca.2.309. Epub 2011 May 25. PMID:21716847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Fossati M., Buzzonetti A., Monego G. Immunological changes in the ascites of cancer patients after intraperitoneal administration of the bispecific antibody catumaxomab (anti-EpCAM × anti-CD3) Gynecol Oncol. August 2015;138(2):343–351. doi: 10.1016/j.ygyno.2015.06.003. Epub 2015 Jun 3. PMID:26049121. [DOI] [PubMed] [Google Scholar]
  • 260.Removab Catumaxomab Product Monograph from Fresnius Biotech. Available at: https://hemonc.org/docs/packageinsert/catumaxomab.pdf.
  • 261.HIGHLIGHTS OF PRESCRIBING INFORMATION: LIBTAYO (cemiplimab-rwlc) injection, for intravenous use Initial U.S. Approval: 09/2018 Last Revised 9/2018 Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761097s000lbl.pdf.
  • 262.Migden M.R., Rischin D., Schmults C.D. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N Engl J Med. July 26, 2018;379(4):341–351. doi: 10.1056/NEJMoa1805131. PMID:29863979. [DOI] [PubMed] [Google Scholar]
  • 263.Lee M.J., Parker C.E., Taylor S.R. Efficacy of medical therapies for fistulizing Crohn's disease: systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2018;16:1879–1892. doi: 10.1016/j.cgh.2018.01.030. Available at: [DOI] [PubMed] [Google Scholar]
  • 264.Trigo-Vicente C., Gimeno-Ballester V., García-López S. Systematic review and network meta-analysis of treatment for moderate-to-severe ulcerative colitis. Int J Clin Pharm. 2018;40:1411–1419. doi: 10.1007/s11096-018-0743-4. Available at: [DOI] [PubMed] [Google Scholar]
  • 265.HIGHLIGHTS OF PRESCRIBING INFORMATION: HUMIRA (adalimumab) injection, for subcutaneous use. Initial U.S. Approval: 2002 Revised 1/2019. Available at: https://www.rxabbvie.com/pdf/humira.pdf.
  • 266.Hanauer S.B., Sandborn W.J., Rutgeerts P. Human anti–tumor necrosis factor monoclonal antibody (adalimumab) in Crohn's disease: the CLASSIC-I trial. Gastroenterology. February 2006;130(2):323–333. doi: 10.1053/j.gastro.2005.11.030. PMID:16472588. [DOI] [PubMed] [Google Scholar]
  • 267.Lee S.D., Rubin D.T., Sandborn W.J. Reinduction with certolizumab pegol in patients with Crohn's disease experiencing disease exacerbation: 7-year Data from the PRECiSE 4 study. Inflamm Bowel Dis. August 2016;22(8):1870–1880. doi: 10.1097/MIB.0000000000000805. PMID:27400222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Winter T.A., Wright J., Ghosh S. Intravenous CDP870, a PEGylated Fab' fragment of a humanized antitumour necrosis factor antibody, in patients with moderate-to severe Crohn's disease: an exploratory study. Aliment Pharmacol Ther. December 2004;20(11–12):1337–1346. doi: 10.1111/j.1365-2036.2004.02285.x. PMID:15606396. [DOI] [PubMed] [Google Scholar]
  • 269.Sandborn W.J., Lee S.D., Randall C. Long-term safety and efficacy of certolizumab pegol in the treatment of Crohn's disease: 7-year results from the PRECiSE 3 study. Aliment Pharmacol Ther. October 2014;40(8):903–916. doi: 10.1111/apt.12930. Epub 2014 Aug 22. PMID:25146586. [DOI] [PubMed] [Google Scholar]
  • 270.Colombel J.F., Sands B.E., Rutgeerts P. The safety of vedolizumab for ulcerative colitis and Crohn's disease. Gut. May 2017;66(5):839–851. doi: 10.1136/gutjnl-2015-311079. Epub 2016 Feb 18. PMID:26893500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Rosario M., Dirks N.L., Milch C. A review of the clinical pharmacokinetics, pharmacodynamics, and immunogenicity of vedolizumab. Clin Pharmacokinet. November 2017;56(11):1287–1301. doi: 10.1007/s40262-017-0546-0. Review. PMID:28523450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Inokuchi T., Takahashi S., Hiraoka S. Long-term outcomes of patients with Crohn's disease who received infliximab or adalimumab as the first-line biologics. J Gastroenterol Hepatol. February 6, 2019 doi: 10.1111/jgh.14624. [Epub ahead of print]. PMID:30724387. [DOI] [PubMed] [Google Scholar]
  • 273.Nelson S.M.L., Nguyen T.M., McDonald J.W.D. Natalizumab for induction of remission in Crohn's disease (Review) Cochrane Database Syst Rev. 2018;(8):CD006097. doi: 10.1002/14651858.CD006097.pub3. PMID:30068022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.HIGHLIGHTS OF PRESCRIBING INFORMATION: TYSABRI (natalizumab) injection, for intravenous use Initial U.S. Approval: 2004 Revised 4/2018. Available at: https://www.tysabri.com/content/dam/commercial/multiple-sclerosis/tysabri/pat/en_us/pdfs/tysabri_prescribing_information.pdf.
  • 275.Lebwohl M., Strober B., Menter A. Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. N Engl J Med. October 2015;373(14):1318–1328. doi: 10.1056/NEJMoa1503824. PMID:26422722. [DOI] [PubMed] [Google Scholar]
  • 276.HIGHLIGHTS OF PRESCRIBING INFORMATION: SILIQ (brodalumab) injection, for subcutaneous use. Initial U.S. Approval: 2017 Revised 2/2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761032lbl.pdf.
  • 277.Blair H.A., Deeks E.D. Abatacept: a review in rheumatoid arthritis. Drugs. July 2017;77(11):1221–1233. doi: 10.1007/s40265-017-0775-4. PMID:28608166. [DOI] [PubMed] [Google Scholar]
  • 278.Goldzweig O., Hashkes P.J. Abatacept in the treatment of polyarticular JIA: development, clinical utility, and place in therapy. Drug Des Dev Ther. January 26, 2011;5:61–70. doi: 10.2147/DDDT.S16489. PMID:21340039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.HIGHLIGHTS OF PRESCRIBING INFORMATION: ORENCIA (abatacept) for injection for intravenous use injection, for subcutaneous use. Initial U.S. Approval: 2005 Revised 12/2013. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125118s171lbl.pdf.
  • 280.Ruiz Garcia V., Burls A., Cabello J.B. Certolizumab pegol (CDP870) for rheumatoid arthritis in adults (Review) Cochrane Database Syst Rev. 2017;(9):CD007649. doi: 10.1002/14651858.CD007649.pub4. PMID:28884785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Navarra S.V., Guzmán R.M., Gallacher A.E. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: a randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377:721–731. doi: 10.1016/S0140-6736(10)61354-2. Epub 2011 Feb 4. PMID:21296403. [DOI] [PubMed] [Google Scholar]
  • 282.Chao Y.S., Adcock L. CADTH; Ottawa: May 2018. CADTH RAPID RESPONSE REPORT: Belimumab Treatment for Adults with Systemic Lupus Erythematosus: A Review of Clinical Effectiveness, Cost-Effectiveness, and Guidelines.https://www.cadth.ca/sites/default/files/pdf/htis/2018/RC0989%20Benlysta%20for%20Lupus%20Final.pdf (CADTH rapid response report: summary with critical appraisal). Publication Date: May 23, 2018. Available at: [PubMed] [Google Scholar]
  • 283.Sciascia S., Radin M., Yazdany J. Efficacy of belimumab on renal outcomes in patients with systemic lupus erythematosus: a systematic review. Autoimmun Rev. March 2017;16 3:287–293. doi: 10.1016/j.autrev.2017.01.010. Epub 2017 Jan 29. PMID:28147262. [DOI] [PubMed] [Google Scholar]
  • 284.Usta C., Turgut N.T., Bedel A. How abciximab might be clinically useful. Int J Cardiol. November 1, 2016;222:1074–1078. doi: 10.1016/j.ijcard.2016.07.213. Epub 2016 Aug 4. Review. PMID:27519521. [DOI] [PubMed] [Google Scholar]
  • 285.Hovingh G.K., Guyton J.R., Langslet G. Alirocumab dosing patterns during 40 months of open-label treatment in patients with heterozygous familial hypercholesterolemia. J Clin Lipidol. 2018 Nov - Dec;12(6):1463–1470. doi: 10.1016/j.jacl.2018.08.011. Epub 2018 Aug 30. PMID:30287210. [DOI] [PubMed] [Google Scholar]
  • 286.Shahawy M.E., Cannon C.P., Blom D.J. Efficacy and safety of alirocumab versus ezetimibe over 2 years (from ODYSSEY COMBO II) Am J Cardiol. September 15, 2017;120(6):931–939. doi: 10.1016/j.amjcard.2017.06.023. Epub 2017 Jun 28. PMID:28750828. [DOI] [PubMed] [Google Scholar]
  • 287.HIGHLIGHTS OF PRESCRIBING INFORMATION: PRALUENT (alirocumab) injection, for subcutaneous use. Initial U.S. Approval: 2015. Revised: 12/2018. Available at: http://products.sanofi.us/praluent/praluent.pdf.
  • 288.Sabatine M.S., Giugliano R.P., Keech A.C. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376:1713–1722. doi: 10.1056/NEJMoa1615664. PMID: 28304224. [DOI] [PubMed] [Google Scholar]
  • 289.HIGHLIGHTS OF PRESCRIBING INFORMATION: REPATHA (evolocumab) injection, for subcutaneous use. Initial U.S. Approval: 2015 Revised 12/2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125522s013lbl.pdf.
  • 290.Kastelein J.J.P., Nissen S.E., Rader D.J. Safety and efficacy of LY3015014, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9): a randomized, placebo-controlled Phase 2 study. Eur Heart J. May 1, 2016;37(17):1360–1369. doi: 10.1093/eurheartj/ehv707. Epub 2016 Jan 12. PMID:26757788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Ridker P.M., Rose L.M., Kastelein J.J.P. Cardiovascular event reduction with PCSK9 inhibition among 1578 patients with familial hypercholesterolemia: results from the SPIRE randomized trials of bococizumab. J Clin Lipidol. 2018 Jul - Aug;12(4):958–965. doi: 10.1016/j.jacl.2018.03.088. Epub 2018 Apr 3. PMID:29685591. [DOI] [PubMed] [Google Scholar]
  • 292.Coles A.J., Cohen J.A., Fox E.J. Alemtuzumab CARE-MS II 5-year follow-up: efficacy and safety findings. Neurology. September 12, 2017;89(11):1117–1126. doi: 10.1212/WNL.0000000000004354. Epub 2017 Aug 23. PMID:28835403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Montalban X., Hauser S.L., Kappos L. Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med. January 19, 2017;376(3):209–220. doi: 10.1056/NEJMoa1606468. PMID:28002688. [DOI] [PubMed] [Google Scholar]
  • 294.Voortman M.M., Greiner P., Moser D. The effect of disease modifying therapies on CD62L expression in multiple sclerosis. Mult Scler J Exp Transl Clin. September 20, 2018;4(3) doi: 10.1177/2055217318800810. 2055217318800810. PMID:30263146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.HIGHLIGHTS OF PRESCRIBING INFORMATION: TYSABRI (natalizumab) injection, for intravenous use. Initial U.S. Approval: 2004. Revised on 1/2012. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/125104s0576lbl.pdf.
  • 296.Guo X., Higgs B.W., Bay-Jensen A.C. Suppression of T cell activation and collagen accumulation by an anti-FNAR1 mAb, Anifrolumab, in adult patients with systemic sclerosis. J Investig Dermatol. October 2015;135(10):2402–2409. doi: 10.1038/jid.2015.188. Epub 2015 May 20. PMID:25993119. [DOI] [PubMed] [Google Scholar]
  • 297.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; A study to assess the safety and efficacy of elezanumab when added to standard of care in relapsing forms of multiple sclerosis. Available at: https://www.clinicaltrials.gov/ct2/show/NCT03737851?term=elezanumab&rank=1.
  • 298.Agius M.A., Klodowska-Duda G., Maciejowski M. Safety and tolerability of inebilizumab (MEDI-551), an anti-CD19 monoclonal antibody, in patients with relapsing forms of multiple sclerosis: results from a phase 1 randomised, placebo-controlled, escalating intravenous and subcutaneous dose study. Mult Scler. February 2019;25(2):235–245. doi: 10.1177/1352458517740641. Epub 2017 Nov 16. PMID:29143550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Constantinescu C.S., Asher A., Fryze W. Randomized phase 1b trial of MOR103, a human antibody to GM-CSF, in multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. May 21, 2015;2(4):e117. doi: 10.1212/NXI.0000000000000117. eCollection 2015 Aug. PMID:26185773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.NIH: National Library of Medicine at Clinicaltrialsgov Clinical trial; A Phase 3, Randomized, Multi-center, Double-blinded, Active-controlled Study to Assess the Efficacy and Safety/Tolerability of Ublituximab (TG-1101; UTX) as Compared to Teriflunomide in Subjects With Relapsing Multiple Sclerosis (RMS) (ULTIMATE 1). Available at: https://www.clinicaltrials.gov/ct2/show/NCT03277261?term=ublituximab&rank=3.
  • 301.Connick P., De Angelis F., Parker R.A. Multiple Sclerosis-Secondary Progressive Multi-Arm Randomisation Trial (MSSMART): a multi-arm phase IIb randomised, double-blind, placebo controlled clinical trial comparing the efficacy of three neuroprotective drugs in secondary progressive multiple sclerosis. BMJ Open. August 30, 2018;8(8):e021944. doi: 10.1136/bmjopen-2018-021944. PMID:30166303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Cadavid D., Balcer L., Galetta S. Safety and efficacy of opicinumab in acute optic neuritis (RENEW): a randomised, placebo-controlled, phase 2 trial. Lancet Neurol. March 2017;16(3):189–199. doi: 10.1016/S1474-4422(16)30377-5. Epub 2017 Feb 15. PMID:28229892. [DOI] [PubMed] [Google Scholar]
  • 303.Bleecker E.R., FitzGerald J.M., Chanez P. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting β2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial. Lancet. October 29, 2016;388(10056):2115–2127. doi: 10.1016/S0140-6736(16)31324-1. Epub 2016 Sep 5. PMID:27609408. [DOI] [PubMed] [Google Scholar]
  • 304.HIGHLIGHTS OF PRESCRIBING INFORMATION: DUPIXENT (dupilumab) injection, for subcutaneous use Initial U.S. Approval: 2017 Last revised: 3/2017.Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761055lbl.pdf.
  • 305.Duggan S. Caplacizumab: first global approval. Drugs. October 2018;78(15):1639–1642. doi: 10.1007/s40265-018-0989-0. Erratum in: Drugs. 2018 Dec;78 (18): 1955. PMID:30298461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Peyvandi F., Scully M., Hovinga J.A.K. Caplacizumab reduces the frequency of major thromboembolic events, exacerbations and death in patients with acquired thrombotic thrombocytopenic purpura. J Thromb Haemost. July 2017;15(7):1448–1452. doi: 10.1111/jth.13716. Epub 2017 Jun 5. PMID:28445600. [DOI] [PubMed] [Google Scholar]
  • 307.Peyvandi F., Scully M., Hovinga J.A.K. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N Engl J Med. February 11, 2016;374(6):511–522. doi: 10.1056/NEJMoa1505533. PMID:26863353. [DOI] [PubMed] [Google Scholar]
  • 308.Scully M., Cataland S.R., Peyvandi F. Caplacizumab treatment for acquired thrombotic thrombocytopenic purpura. N Engl J Med. January 24, 2019;380(4):335–346. doi: 10.1056/NEJMoa1806311. Epub 2019 Jan 9. PMID:30625070. [DOI] [PubMed] [Google Scholar]
  • 309.Ataga K.I., Kutlar A., Kanter J. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. February 2, 2017;376(5):429–439. doi: 10.1056/NEJMoa1611770. Epub 2016 Dec 3. PMID:27959701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; Study of Dose Confirmation and Safety of Crizanlizumab in Pediatric Sickle Cell Disease Patients. Available at: https://clinicaltrials.gov/ct2/show/NCT03474965?term=crizanlizumab&rank=1.
  • 311.Johnson S., Gerding D.N. Bezlotoxumab. Clin Infect Dis. February 1, 2019;68(4):699–704. doi: 10.1093/cid/ciy577. PMID:30020417. [DOI] [PubMed] [Google Scholar]
  • 312.Navalkele B.D., Chopra T. Bezlotoxumab: an emerging monoclonal antibody therapy for prevention of recurrent Clostridium difficile infection. Biologics. January 18, 2018;12:11–21. doi: 10.2147/BTT.S127099. eCollection 2018. Review. PMID:29403263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Simões E.A.F., Bont L., Manzoni P. Past, present and future approaches to the prevention and treatment of respiratory syncytial virus infection in children. Infect Dis Ther. March 2018;7(1):87–120. doi: 10.1007/s40121-018-0188-z. Epub 2018 Feb 22. Review. PMID:29470837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Tripp R.A., Power U.F., Openshaw P.J.M. Respiratory syncytial virus: targeting the G protein provides a new approach for an old problem. J Virol. January 17, 2018;92(3):e01302–e01317. doi: 10.1128/JVI.01302-17. Print 2018 Feb 1. PMID:29118126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.NIH: National Library of Medicine at Clinicaltrials.gov Clinical trial; 155 Studies found for: basiliximab. Available at: https://clinicaltrials.gov/ct2/results?cond=&term=basiliximab&cntry=&state=&city=&dist= .
  • 316.Sun Z.J., Du X., Su L.L. Efficacy and safety of basiliximab versus daclizumab in kidney transplantation: a meta-analysis. Transplant Proc. October 2015;47(8):2439–2445. doi: 10.1016/j.transproceed.2015.08.009. PMID: 26518947. [DOI] [PubMed] [Google Scholar]
  • 317.Rostainga L., Salibab F., Calmusc Y. Review article: use of induction therapy in liver transplantation. Transplant Rev. October 2012;26(4):246–260. doi: 10.1016/j.trre.2012.06.002. Epub 2012 Aug 3. Review. PMID:22863028. [DOI] [PubMed] [Google Scholar]
  • 318.Kittipibul V., Tantrachoti P., Ongcharit P. Low- dose basiliximab induction therapy in heart transplantation. Clin Transplant. December 2017;31(12) doi: 10.1111/ctr.13132. Epub 2017 Nov 3. PMID:28990220. [DOI] [PubMed] [Google Scholar]
  • 319.Butts R.J., Dipchand A.I., Sutcliffe D. Comparison of basiliximab vs antithymocyte globulin for induction in pediatric heart transplant recipients: an analysis of the International Society for Heart and Lung Transplantation database. Pediatr Transplant. June 2018;22(4):e13190. doi: 10.1111/petr.13190. PMID:29878688. [DOI] [PubMed] [Google Scholar]
  • 320.Kutilek S. Burosumab: a new drug to treat hypophosphatemic rickets. Sudan J Paediatr. 2017;17(2):71–73. doi: 10.24911/SJP.2017.2.11. PMID:29545670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Insogna K.L., Briot K., Imel E.A. A randomized, double-blind, placebo-controlled, phase 3 trial evaluating the efficacy of burosumab, an anti-FGF23 antibody, in adults with X-linked hypophosphatemia: week 24 primary analysis. J Bone Miner Res. August 2018;33(8):1383–1393. doi: 10.1002/jbmr.3475. Epub 2018 Jun 26. PMID:29947083. [DOI] [PubMed] [Google Scholar]
  • 322.Cummings S.R., San Martin J M.D., McClung M.R. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. August 20, 2009;361(8):756–765. doi: 10.1056/NEJMoa0809493. Epub 2009 Aug 11. Erratum in: N Engl J Med. 2009 Nov 5; 361(19): 1914. PMID:19671655. [DOI] [PubMed] [Google Scholar]
  • 323.Zaheer S., LeBoff M., Lewiecki E.M. Denosumab for the treatment of osteoporosis. Expert Opin Drug Metabol Toxicol. March 2015;11(3):461–470. doi: 10.1517/17425255.2015.1000860. Epub 2015 Jan 22. PMID:25614274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Deeks E.D. Denosumab: a review in postmenopausal osteoporosis. Drugs Aging. February 2018;35(2):163–173. doi: 10.1007/s40266-018-0525-7. Review. Erratum in: Drugs Aging. 2018 Mar 9. PMID:29435849. [DOI] [PubMed] [Google Scholar]
  • 325.Gül G., Sendur M.A.N., Aksoy S. A comprehensive review of denosumab for bone metastasis in patients with solid tumors. Curr Med Res Opin. 2016;32(1):133–145. doi: 10.1185/03007995.2015.1105795. Epub 2015 Nov 25. Review. PMID:26451465. [DOI] [PubMed] [Google Scholar]
  • 326.Raje N., Terpos E., Willenbacher W. Denosumab versus zoledronic acid in bone disease treatment of newly diagnosed multiple myeloma: an international, double-blind, double-dummy, randomised, controlled, phase 3 study. Lancet Oncol. March 2018;19(3):370–381. doi: 10.1016/S1470-2045(18)30072-X. Epub 2018 Feb 9. PMID:29429912. [DOI] [PubMed] [Google Scholar]
  • 327.Dugel P.U., Jaffe G.J., Sallstig P. Brolucizumab versus Aflibercept in participants with neovascular age-related macular degeneration: a randomized trial. Ophthalmology. September 2017;124(9):1296–1304. doi: 10.1016/j.ophtha.2017.03.057. Epub 2017 May 24. PMID:28551167. [DOI] [PubMed] [Google Scholar]
  • 328.HIGHLIGHTS OF PRESCRIBING INFORMATION: ILARIS (canakinumab) injection, for subcutaneous use. Initial U.S. Approval: 2009. Last revised 12/2016. Available at: https://www.pharma.us.novartis.com/sites/www.pharma.us.novartis.com/files/ilaris.pdf.
  • 329.Katoh M. Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (Review) Int J Mol Med. August 2018;42(2):713–725. doi: 10.3892/ijmm.2018.3689. Epub 2018 May 17. PMID:29786110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.KATOH M., Katoh M. Molecular genetics and targeted therapy of WNT-related human diseases (Review) Int J Mol Med. September 2017;40(3):587–606. doi: 10.3892/ijmm.2017.3071. Epub 2017 Jul 19. PMID:28731148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Department of Health and Human Services . January 2002. Food and Drug Administration, Center for Biologics Evaluation and Research (CBER). Guidance for Industry. Revised Preventive Measures to Reduce the Possible Risk of Transmission of Creutzfeldt-Jacob Disease (CJD) and Variant Creutzfeldt-Jacob Disease (vCJD) by Blood and Blood Products. From the US Food and Drug Administration (FDA) website. [Google Scholar]

Further reading

  • 1.Centers for Disease Control Immunization Practices Advisory Committee (ACIP) Diphtheria, tetanus, and pertussis: recommendations for vaccine use and other preventive measures. MMWR Recomm Rep (Morb Mortal Wkly Rep) 1991;40(RR-10):1–28. [PubMed] [Google Scholar]
  • 2.Centers for Disease Control Update on adult immunization: recommendations of the immunization practices advisory committee (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 1991;40(RR-12) 18,49,70,87. [PubMed] [Google Scholar]
  • 3.Kretsinger K., Broder K.R., Cortese M.M. Preventing tetanus, diphtheria, and pertussis among adults: use of tetanus toxoid, reduced diphtheria toxoid and acellular pertussis vaccine recommendations of the Advisory Committee on Immunization Practices (ACIP) and recommendation of ACIP, supported by the Healthcare Infection Control Practices Advisory Committee (HICPAC), for use of Tdap among health-care personnel. MMWR Recomm Rep (Morb Mortal Wkly Rep) 2006;55:1–37. [PubMed] [Google Scholar]
  • 4.Broder K.R., Cortese M.M., Iskander J.K. Preventing tetanus, diphtheria, and pertussis among adolescents: use of tetanus toxoid, reduced diphtheria toxoid and acellular pertussis vaccines recommendations of the Advisory Committee on Immunization Practices (ACIP) MMWR Recomm Rep (Morb Mortal Wkly Rep) 2006;55:1–34. [PubMed] [Google Scholar]
  • 5.Snydman D.R., Werner B.G., Dougherty N.N. Cytomegalovirus immune globulin prophylaxis in liver transplantation: a randomized, double-blind, placebo-controlled trial. Ann Intern Med. 1993;119:984–991. doi: 10.7326/0003-4819-119-10-199311150-00004. [DOI] [PubMed] [Google Scholar]
  • 6.Bowden R.A., Sayers M., Flournoy N. Cytomegalovirus immune globulin and seronegative blood products to prevent primary cytomegalovirus infection after marrow transplantation. N Engl J Med. 1986;314:1006–1010. doi: 10.1056/NEJM198604173141602. [DOI] [PubMed] [Google Scholar]
  • 7.Mair-Jenkins J., Saavedra-Campos M., Baillie J.K. The effectiveness of convalescent plasma and hyperimmune immunoglobulin for the treatment of severe acute respiratory infections of viral etiology: a systematic review and exploratory meta-analysis. J Infect Dis. 2015;211:80–90. doi: 10.1093/infdis/jiu396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.US Food and Drug Administration . US Food and Drug Administration; Silver Spring, MD: 2011. Guidance for Industry Influenza: Developing Drugs for Treatment And/or Prophylaxis. [Google Scholar]
  • 9.Ohmit S.E., Petrie J.G., Cross R.T., Johnson E., Monto A.S. Influenza hemagglutination-inhibition antibody titer as a correlate of vaccine-induced protection. J Infect Dis. 2011;204:1879–1885. doi: 10.1093/infdis/jir661. [DOI] [PubMed] [Google Scholar]
  • 10.Arnon S.S., Schechter R., Inglesby T.V. for the Working Group on Civilian Biodefense. Botulinum toxin as a biologic weapon: medical and public health management. J Am Med Assoc. 2001;285:1059–1070. doi: 10.1001/jama.285.8.1059. [DOI] [PubMed] [Google Scholar]

Articles from Immunologic Concepts in Transfusion Medicine are provided here courtesy of Elsevier

RESOURCES