13
Immunotoxin Therapy of Hematologic Malignancies Arthur E. Frankel, David M. Neville, Thomas A. Bugge, Robert J. Kreitman, and Stephen H. Leppla Patients with chemotherapy relapsed or refractory he- matologic malignancies may be effectively treated with allogeneic or autologous stem cell transplants. How- ever, many patients cannot be transplanted due to age, comorbidities, or lack of suitable donors. Further, a fraction of patients relapse post-transplant. Novel ther- apeutic agents that can kill multidrug-resistant malig- nant stem cells and are not myelosuppressive are needed. One class of such agents is immunotoxins. Immunotoxins consist of cell-selective ligands co- valently linked to peptide toxins. The ligand delivers the molecule to specific cell surface receptors on ma- lignant cells. The toxin triggers cell death either by reaching the cytosol and catalytically inactivating vital cell processes or by modifying the tumor cell surface membrane. We have synthesized immunotoxins for therapy of chemoresistant hematologic diseases. In this review, we will detail the synthesis of a number of these drugs and describe their preclinical and clinical activity. Several of these agents have shown dramatic antitumor effects in patients with hematologic neo- plasms, and one immunotoxin has been approved for use by the US Food and Drug Administration (FDA). Over the next several decades, a growing number of these agents should reach the clinic. Semin Oncol 30:545-557. © 2003 Elsevier Inc. All rights reserved. P ATIENTS WITH CHEMOTHERAPY re- lapsed or refractory hematologic malignancies may be effectively treated with allogeneic or au- tologous stem cell transplants. 1 However, many patients cannot be transplanted due to age, comor- bidities, or lack of suitable donors. Further, a frac- tion of patients relapse post-transplant. Novel therapeutic agents that can kill multidrug-resistant malignant stem cells and are not myelosuppressive are needed. One class of such agents is immuno- toxins. Immunotoxins consist of cell-selective li- gands covalently linked to peptide toxins. The ligand delivers the molecule to specific cell surface receptors on malignant cells. The toxin triggers cell death either by reaching the cytosol and cat- alytically inactivating vital cell processes or by modifying the tumor cell surface membrane. We have synthesized immunotoxins for therapy of chemoresistant hematologic diseases. In this re- view, we will detail the synthesis of a number of these drugs and describe their preclinical and clin- ical activity. Several of these agents have shown dramatic antitumor effects in patients with hema- tologic neoplasms, and one immunotoxin has been approved for use by the US Food and Drug Ad- ministration (FDA). Over the next several de- cades, a growing number of these agents should reach the clinic. SYNTHESIS OF IMMUNOTOXINS Monoclonal antibodies, antibody fragments, and cytokines reactive with cell surface antigens and receptors with restricted expression to human hematopoietic cells have been used in immuno- toxin synthesis. The cell surface targets have in- cluded the cluster designation differentiation an- tigens: CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD10, CD19, CD22, CD24, CD25, CD26, CD29, CD30, CD33, CD37, CD38, CD45, CD45RO, CD52, CD54, CD56, CD64, CD72, CD80, CD86, and CD122. Additional cell surface targets include the cytokine receptors: interleukin-2 receptor (IL- 2R), granulocyte-macrophage colony-stimulating factor receptor (GM-CSFR), interleukin-3 recep- tor (IL-3R), interleukin-4 receptor (IL-4R), inter- leukin-6 receptor (IL-6R), interleukin-7 receptor (IL-7R), interleukin-9 receptor (IL-9R), interleu- kin-13 receptor (IL-13R), granulocyte colony- stimulating factor receptor (G-CSFR), urokinase receptor (uPAR), transferrin receptor (TfR), and interleukin-15 receptor (IL-15R). Finally, there is a heterogeneous group of hematopoietic receptors, including CCR5, human leukocyte antigen (HLA)- DR, immunoglobulin (Ig) idiotype, IgMFc, the CLL2m chronic lymphocytic leukemia antigen, the ML30 heat shock protein, the kappa-myeloma antigen (KMA) lymphoblastoid antigen, the M5 acute myeloid leukemia (AML) antigen, the HAE9 and HAE3 erythroblast antigens, the SN7 B-cell antigen, the 8A and 62B1 myeloma anti- gens, neurokinin-1, and the NDA4, IORT6, and SOKT1 T-cell antigens. Not all ligand-toxins From the Wake Forest University School of Medicine, Winston- Salem, NC; National Institute of Mental Health, Bethesda, MD; National Institute of Dental and Craniofacial Research, Bethesda, MD; National Cancer Institute, Bethesda, MD; National Institute of Allergy and Infectious Diseases, Bethesda, MD. Address reprint requests Arthur E. Frankel, MD, Hanes 406, Wake Forest University of School, Medical School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157. © 2003 Elsevier Inc. All rights reserved. 0093-7754/03/3004-0008$30.00/0 doi:10.1016/S0093-7754(03)00241-0 545 Seminars in Oncology, Vol 30, No 4 (August), 2003: pp 545-557

Immunotoxin therapy of hematologic malignancies

Embed Size (px)

Citation preview

Page 1: Immunotoxin therapy of hematologic malignancies

Immunotoxin Therapy of Hematologic Malignancies

Arthur E. Frankel, David M. Neville, Thomas A. Bugge, Robert J. Kreitman, and Stephen H. Leppla

Patients with chemotherapy relapsed or refractory he-matologic malignancies may be effectively treated withallogeneic or autologous stem cell transplants. How-ever, many patients cannot be transplanted due to age,comorbidities, or lack of suitable donors. Further, afraction of patients relapse post-transplant. Novel ther-apeutic agents that can kill multidrug-resistant malig-nant stem cells and are not myelosuppressive areneeded. One class of such agents is immunotoxins.Immunotoxins consist of cell-selective ligands co-valently linked to peptide toxins. The ligand deliversthe molecule to specific cell surface receptors on ma-lignant cells. The toxin triggers cell death either byreaching the cytosol and catalytically inactivating vitalcell processes or by modifying the tumor cell surfacemembrane. We have synthesized immunotoxins fortherapy of chemoresistant hematologic diseases. Inthis review, we will detail the synthesis of a number ofthese drugs and describe their preclinical and clinicalactivity. Several of these agents have shown dramaticantitumor effects in patients with hematologic neo-plasms, and one immunotoxin has been approved foruse by the US Food and Drug Administration (FDA).Over the next several decades, a growing number ofthese agents should reach the clinic.Semin Oncol 30:545-557. © 2003 Elsevier Inc. All rightsreserved.

PATIENTS WITH CHEMOTHERAPY re-lapsed or refractory hematologic malignancies

may be effectively treated with allogeneic or au-tologous stem cell transplants.1 However, manypatients cannot be transplanted due to age, comor-bidities, or lack of suitable donors. Further, a frac-tion of patients relapse post-transplant. Noveltherapeutic agents that can kill multidrug-resistantmalignant stem cells and are not myelosuppressiveare needed. One class of such agents is immuno-toxins. Immunotoxins consist of cell-selective li-gands covalently linked to peptide toxins. Theligand delivers the molecule to specific cell surfacereceptors on malignant cells. The toxin triggerscell death either by reaching the cytosol and cat-alytically inactivating vital cell processes or bymodifying the tumor cell surface membrane. Wehave synthesized immunotoxins for therapy ofchemoresistant hematologic diseases. In this re-view, we will detail the synthesis of a number ofthese drugs and describe their preclinical and clin-ical activity. Several of these agents have showndramatic antitumor effects in patients with hema-tologic neoplasms, and one immunotoxin has beenapproved for use by the US Food and Drug Ad-

ministration (FDA). Over the next several de-cades, a growing number of these agents shouldreach the clinic.

SYNTHESIS OF IMMUNOTOXINS

Monoclonal antibodies, antibody fragments,and cytokines reactive with cell surface antigensand receptors with restricted expression to humanhematopoietic cells have been used in immuno-toxin synthesis. The cell surface targets have in-cluded the cluster designation differentiation an-tigens: CD2, CD3, CD4, CD5, CD6, CD7, CD8,CD10, CD19, CD22, CD24, CD25, CD26, CD29,CD30, CD33, CD37, CD38, CD45, CD45RO,CD52, CD54, CD56, CD64, CD72, CD80, CD86,and CD122. Additional cell surface targets includethe cytokine receptors: interleukin-2 receptor (IL-2R), granulocyte-macrophage colony-stimulatingfactor receptor (GM-CSFR), interleukin-3 recep-tor (IL-3R), interleukin-4 receptor (IL-4R), inter-leukin-6 receptor (IL-6R), interleukin-7 receptor(IL-7R), interleukin-9 receptor (IL-9R), interleu-kin-13 receptor (IL-13R), granulocyte colony-stimulating factor receptor (G-CSFR), urokinasereceptor (uPAR), transferrin receptor (TfR), andinterleukin-15 receptor (IL-15R). Finally, there is aheterogeneous group of hematopoietic receptors,including CCR5, human leukocyte antigen (HLA)-DR, immunoglobulin (Ig) idiotype, IgMFc, theCLL2m chronic lymphocytic leukemia antigen,the ML30 heat shock protein, the kappa-myelomaantigen (KMA) lymphoblastoid antigen, the M5acute myeloid leukemia (AML) antigen, theHAE9 and HAE3 erythroblast antigens, the SN7B-cell antigen, the 8A and 62B1 myeloma anti-gens, neurokinin-1, and the NDA4, IORT6, andSOKT1 T-cell antigens. Not all ligand-toxins

From the Wake Forest University School of Medicine, Winston-Salem, NC; National Institute of Mental Health, Bethesda, MD;National Institute of Dental and Craniofacial Research, Bethesda,MD; National Cancer Institute, Bethesda, MD; National Instituteof Allergy and Infectious Diseases, Bethesda, MD.

Address reprint requests Arthur E. Frankel, MD, Hanes 406,Wake Forest University of School, Medical School of Medicine,Medical Center Blvd, Winston-Salem, NC 27157.

© 2003 Elsevier Inc. All rights reserved.0093-7754/03/3004-0008$30.00/0doi:10.1016/S0093-7754(03)00241-0

545Seminars in Oncology, Vol 30, No 4 (August), 2003: pp 545-557

Page 2: Immunotoxin therapy of hematologic malignancies

Table 1. In Vitro Potency of Immunotoxins for Hematologic Malignancies

Agent Receptor Normal Tissue IC50 (pmol/L) Reference

Directed at CD AntigensOKT11-saporin CD2 T cells 0.5 2A-dmDT390-bisFv(G4S) CD3 T cells 0.2 3TEC-T4-saporin CD4 T cells 1,000 4H65-rGeloninD247C CD5 T cells, rare B cells 41 5Anti–CD6-blocked ricin CD6 Thymocytes 4 8BA1e-deglycosylated ricin A CD7 T cells 5 9WT82-ricin A CD8 T cells �10,000 10SN5-ricin A CD10 Granulocytes, fibroblasts 2,000 11My7/Fab�-GAMIg-ricin A CD13 Granulocytes, monocytes �10,000 201G10/Fab�-GAMIg-ricin A CD15 Granulocytes, monocytes 5,000 20BU12-saporin CD19 B cells, dendritic cells 35 12RTB4(Fv)100THW100A-PE38 CD22 B cells 16 13SWAII-ricin A CD24 B cells, granulocytes 70 25Anti–Tac(Fv)-PE38 CD25 T cells, B cells, macrophages 2 141F7-blocked ricin CD26 T cells �1,000 154B4-blocked ricin CD29 Lymphocytes, endothelium 500 16T25(dsFv)-PE38 CD30 T cells, B cells 160 17HuM195-rgelonin CD33 Monocytes, granulocytes 50 18MB-1/anti-saporin/saporin CD37 T cells, B cells, myeloid �30,000 19OKT10-saporin CD38 Lymphocytes, macrophages 38 21G28-5sFv-bryodin 1 CD40 B cells, dendritic cells 100 22BMAC1-ricin A CD45 Leukocytes 3,000 23UCHL1-ricin A CD45RO Leukocytes 1,000 24Campath-1-saporin CD52 Leukocytes 200 26UV3-deglycosylated ricin A CD54 Granulocytes, monocytes 40 27N901-blocked ricin CD56 NK cells, neural tissues 50 28H22-deglycosylated ricin A CD64 Monocytes, macrophages 4,000 29J3-109-PAP CD72 B cells 100 30M24-saporin CD80 B cells, macrophages 0.3 311G10-saporin CD86 Monocytes, dendritic cells 3 31Mik-beta1(Fv)-PE40 CD122 Leukocytes 100 32

Directed at Cytokine ReceptorsDAB389IL2 IL-2R Activated T, B cells 4 33DT388IL3 IL-3R Mast cells, myeloid stem 15 34DAB389IL4 IL-4R Hepatocytes, endothelium 50 35IL6-PE664Glu IL-6R Hepatocytes 30 36DAB389IL7 IL-7R Thymocytes, B cells, T cells 100 37rhIL9-ETA� IL-9R Thymocytes, mast cells 6,000 38IL13-PE38QQR IL-13R Skin, lung, heart, liver cells 0.8 39DAB389IL15 IL-15R Leukocytes, skin, lung, liver 7,000 40GCSF-PE40 G-CSFR Granulocytes 100 41DT388GMCSF GM-CSFR Granulocytes, macrophages 3 42DTAT uPAR Endothelium 20 43

Directed at Miscellaneous AntigensRANTES-PE38 CCR5 T cells �5,000 44DAB389SP Neurokinin-1 Neurons, endothelium 18 452G5-ricin A HLA-DR Bcells, T cell, dendritic �10,000 4638-13-ricin A Ceramide TH Pk1/Pk2 cells 200 47CLL2m-ricin A CLL2m antigen ND 23 48HAE3-ricin A Glycophorin RBC 3,100 49HAE9-ricin A Erythroid antigen Erythroblasts 320 49TEC-saporin IgM B cells 100 50IOR-T6-HT Thymocyte antigen Thymocytes 5,000 518A-saporin 8A myeloma antigen B cells 60 5262B1-saporin 62Bmyeloma antigen B cells 1 52

546 FRANKEL ET AL

Page 3: Immunotoxin therapy of hematologic malignancies

have shown potent selective toxicity. In somecases, the ligand-receptor is expressed at very lowlevels on the malignant cells or is poorly internal-ized (CD4, CD8, CD10, CD13, CD15, CD26,CD37, CD45, glycophorin, HLA-DR, IL-9R, andIL-15R). In other cases, receptor is present oncritical normal tissues (TfR, IL-4R and IL-6R).Table 1 shows a list of the immunotoxins synthe-sized for therapy of human hematologic cancers,important normal tissues with the targeted recep-tor when present, and a representative concentra-tion of the agent reducing malignant cell viabilityby 50% (IC50). In addition to receptor specificity,other ligand variables in immunotoxin potencyinclude the affinity, distance of the antigen/recep-tor epitope from the cell membrane, and size andconformation of the ligand.6,7

All the toxins used in immunotoxin synthesisfor treatment of hematologic tumors have beencytotoxins. Cytotoxins can directly induce celldeath. There are three classes of peptide cytotox-ins, and each has been used. Class I toxins areintracellular enzymes; class II proteins bind to cellsurfaces and trigger intracellular signal pathways;and class III cytotoxins are pore-forming peptidesthat cause leaks in the plasma membrane. Most ofthe immunotoxins used for hematologic malignan-cies have been made with class I cytotoxins.

The exceptions that have not used class I cyto-toxins include the class II recombinant cytotoxinconjugate, anti-Tac(Fab)-PLC, and the class III

cytotoxin immunoconjugates: IOR-T6-HT, ScFv-mel-FLAG, and OKT1-PT . The class II immuno-toxin consists of the Fab fragment of the anti-CD25 monoclonal antibody (anti-Tac) fused tophospholipase C.57 This conjugate reacts with theIL-2R �-subunit (CD25) on activated or malig-nant T and B cells. The molecule deliversphospholipase C to the surface of the malignantlymphocyte, which in turn hydrolyzes phosphati-dylcholine triggering abnormal cell signaling anddeath. The conjugate had an IC50 of 20 pmol/L.The class III cytotoxin conjugates—IOR-T6-HT,ScFv-mel-FLAG, OKT1-PT, and T3-3A1-CTX—have been studied in vitro. IOR-T6-HT binds to aT-cell differentiation antigen on malignant T cellsand the hemolytic toxin creates pores in the mem-brane.51 ScFv-mel-FLAG binds the KMA antigenon human myeloma cells where the associatedinsect class III toxin, mellitin amphipathic pep-tide, punches holes in the myeloma cell mem-brane.58 OKT1-PT consists of the anti-CD5 anti-body OKT1 coupled to the plant Pyrularia thioninmembrane active peptide.59 Again, delivery of themolecule to T-cell surfaces is followed by mem-brane pore formation and leakage. Finally, T3-3A1-CTX consists of an anti–T-cell monoclonalantibody coupled to the cobra venom cationic 63amino acid membrane lytic peptide toxin.60 Un-fortunately, there is no information on the in vivosafety of any of the class II and class III cytotoxinconjugates. Since all of these toxins may react

Table 1. In Vitro Potency of Immunotoxins for Hematologic Malignancies (Contd.)

Agent Receptor Normal Tissue IC50 (pmol/L) Reference

Directed at Miscellaneous Antigens (Cont’d)SN7-RTA SN7 B cell antigen B cells 2 53LAM3-saporin M5b-AML antigen Monocytes, platelets 70 54NDA4-gelonin NDA4 T-cell antigen T cells, B cells 200 55ML30-saporin HSP65 Virus infected cells 1,000 56

NOTE. Cell lines, toxin used, and duration of incubation vary, which makes comparisons of different ligands difficult. Normal tissue reactivityaffected by toxin. In the case of blocked ricin, residual galactosyl binding causes endothelial reactivity and toxicity and may contribute to tumorcell killing.

Abbreviations: rgelonin, recombinant gelonin; PE, Pseudomonas exotoxin; PE38, 38-kd fragment of PE; PE40, 40-kd fragment of PE; PAP,pokeweed antiviral protein; DT, diphtheria toxin; DAB389, DT amino acids residues 1-389; DT388, DT amino acid residues 1-388; PE664Glu,PE-modified with 4 lysines changed to glutamic acids to reduce normal tissue binding; ETA; mutant PE with reduced normal tissue binding;PE38QQR, modified 38-kd PE fragment; uPAR, urokinase-plasminogen-activator receptor; G-CSF, granulocyte colony-stimulating factor;GM-CSF, granulocyte-macrophage colony-stimulating factor; DTAT, diphtheria toxin fragment fused to the N-terminal fragment of urokinase;TH, trihexoside; RBC, red blood cells, Pk1/Pk2, rare Pk blood group allotypes; HT, hemolytic toxin; HSP65, heat shock protein 65 kd Mr; ND,not determined.

IMMUNOTOXIN THERAPY OF HEMATOLOGIC MALIGNANCIES 547

Page 4: Immunotoxin therapy of hematologic malignancies

with cell surface membranes of blood cells andendothelium, it will be important to determine thein vivo safety of these immunotoxins.

The class I toxin conjugates offer additionalsafety, since they only damage cells after ligand-receptor internalization. Most of the employedclass I toxins inactivate cytosolic protein synthesiseither by modifying elongation factor 2 (diphthe-ria toxin [DT} and Pseudomonas exotoxin [PE]) orby degrading ribosomal RNA (ricin, abrin, vis-cumin, gelonin, saporin) or total RNA (ribonucle-ase, angiogenin,). Bax triggers mitochondrial-me-diated apoptosis. Some of the type I cytotoxinshave normal tissue binding sites, which must beremoved or modified prior to conjugation. Thesetoxins include ricin, abrin, viscumin, DT, and PE.A large group of cytotoxins do not have normaltissue binding sites and must be linked to cellsurface reactive ligands to intoxicate cells. Thesetoxins include gelonin, saporin, pokeweed antivi-ral protein (PAP), bryodin, bouganin, momordin,

dianthin, momorcochin, trichokirin, luffin, restric-tocin, mitogillin, �-sarcin, Onconase, pancreaticribonuclease, Bax, eosinophil-derived neurotoxin,and angiogenin. Examples of the class I toxins usedin hematologic cancer immunotoxins are shown inTable 2. Which toxins provide the greatest spec-ificity and potency? Complete inactivation of thenormal tissue binding sites is necessary. Partialinactivation, as was observed with the blockedricin conjugates, yields a poor therapeutic index.Direct comparisons of immunotoxins targeting thesame antigen/receptor on nonmyeloid cells withdifferent toxins in which the normal tissue bindingsites have been eliminated have been made withimmunotoxins reactive with CD2, CD3, CD5,CD7, CD19, CD22, CD25, CD30, IL-2R, GM-CSFR, IL-4R, IL-6R, uPAR, IL-13R, and TfR.The anti-TfR immunoconjugates include the mostmembers for comparisons among toxins (Table 3).The best cytotoxins for conjugate synthesis in-clude the bacterial holotoxins—DT and PE—in

Table 2. Peptide Class I Cytotoxins Used in Hematologic Neoplasm Immunotoxins

Conjugate Toxin Receptor IC50 (pmol/L) Reference

A-dmDT390-bisFv(G4S) DT CD3 0.2 3RTB4(Fv)100THW100A-PE38 PE CD22 16 133A1e-deglycosylated ricin A Ricin CD7 5 9Anti-CD25-MLA Viscumin CD25 400 61M24-bouganin Bouganin CD80 10 31BU12-saporin Saporin CD19 35 12H65-rGeloninD247C Gelonin CD5 41 5G28-5sFv-bryodin 1 Bryodin CD40 100 22J3-109-PAP PAP CD72 100 308A-momordin Momordin 8A myeloma antigen 200 62BerH2-rdianthin 30 Dianthin 30 CD30 100 63UCHT1/anti-Igdianthin 32 Dianthin 32 CD3 10 64UCHT1/anti-Igmomorcochin Momorcochin CD3 68 64UCHT1/anti-Igtrichokirin Trichokirin CD3 32 64HB21-luffin Luffin TfR 50 79RSFv Restrictocin TfR 40 72H65-MS-mitogillin Mitogillin CD5 150 65HB21-�-sarcin �-sarcin TfR 30 80LL2-onconase Onconase CD22 20 66T101-ribonuclease A RibonucleaseA CD5 120,000 67IL2-Bax Bax IL2R 140,000 68hpRNase1-IL2 hpRNase 1 IL2R 20,000 69EDNsFv EDN TfR 500 70AngFBsFvL2 Angiogenin TfR 5,000 71Tf-ribonuclcase Ribonuclease TfR 100,000 73

Abbreviations: rdianthin, recombinant dianthin; hpRNase, human pancreatic ribonuclease; EDN, eosinophil-derived neurotoxin; TfR, trans-ferrin receptor; Tf, transferrin.

548 FRANKEL ET AL

Page 5: Immunotoxin therapy of hematologic malignancies

which the binding domains have been selectivelymodified with preservation of the translocationdomains. These conjugates show similar potencyin non-myeloid cells. The immunotoxins withplant ribosome-inactivating proteins—ricin Achain, viscumin A chain, gelonin, saporin,pokeweed antiviral protein, bryodin, bouganin,momordin, dianthin, luffin, momorcochin, and tri-chokirin—have intermediate potency. The mech-anism by which they reach the cytosol remainsunknown. Immunotoxins with the fungal nucle-ases—restrictocin, mitogillin, and �-sarcin—alsohave intermediate affinity, The amphibian, bo-vine, and human nucleases—onconase, ribonucle-ase, eosinophil-derived neurotoxin, angiogeninand the human pro-apoptotic protein Bax—havethe least potency. Immunotoxins for myeloid leu-kemias are a special case. Because of the rapidtrafficking of endosomes to lysosomes and the re-duced routing to the Golgi and endoplasmic retic-ulum in myeloid cells, DT conjugates work best.74

DT can escape to the cytosol from early endo-somes to the cytosol, while PE and ricin toxin onlyreach the cytosol after passage through the Golgiand endoplasmic reticulum. Another factor influ-encing efficacy is immunogenicity. Patients withantitoxin antibodies clear immunotoxins rapidlyfrom the bloodstream and have a marked reducedarea under the curve (AUC). Since most peopleare immunized with DT, there is a significant

pretreatment antibody titer in the blood of manypatients and an amnestic response occurs in addi-tional patients treated with DT conjugates. Ofintermediate immunogenicity are the toxins thatare foreign antigens to which people have notbeen previously exposed. These toxin conjugatesevoke antibodies after a lag of 3 to 4 weeks. Theseinclude most of the cytotoxins with the exceptionof human proteins—Bax, ribonuclease, eosinophilneurotoxin, and angiogenin. Since these toxinshave the least potency, selection of the cytotoxinfor a particular application involves compromisesbetween immunogenicity, potency, and specificity.Pharmacologic problems due to the toxin havebeen observed in vivo. Vascular leak syndromefrom vascular injury may be due to selective re-ceptors for peptide toxins on endothelial cells.Modification of the endothelial binding residueson toxins may reduce this toxicity and has beentested on ricin A chain conjugates.75 Some of thecytotoxin conjugates are small, less than 50 kd Mr.Such conjugates may be rapidly cleared by thekidney glomeruli leading to a short half-life, poorAUC, and renal tubular injury. One approach toovercome this problem is to enlarge the size of theconjugate so that glomerular filtration is reduced.This has been successfully accomplished with sev-eral anti-CD3 recombinant DT fusion proteinsleading to less renal toxicity and improvedAUC.3,76 Another approach to reduce the nontar-get tissue toxicities of immunotoxins is to addanother tissue-specific requirement for cell intox-ication. Since several bacterial toxins requirefurin-mediated cleavage prior to translocation ofthe catalytic domain to the cytosol, replacing thefurin sequences with sequences specific to tumor-selective proteases would provide additional spec-ificity. This has been accomplished for anthraxprotective antigen in combination with a fusionmolecule of the N-terminal fragment of lethalfactor with the translocation and catalytic do-mains of PE (FP59).77,78 The resulting toxins showtumor-selective cytotoxicity and safety in animals.Similar modifications should be possible with DTand PE conjugates. Also important in immuno-toxin design is the choice of linker between theligand and toxin.

The covalent attachment of the toxin and li-gand has to meet several requirements. It must bestable in the bloodstream and interstitial fluid. Itshould not impair ligand affinity to its receptor,and it must not impede toxin translocation or

Table 3. Comparison of the Potency of Anti-transferrin Receptor Immunoconjugates

Immunotoxin ToxinIC50

(pmol/L) Reference

DT388anti-TfR(Fv) DT 3 85Anti-TfR(Fv)PE38 PE 6 85454A12-ricin A Ricin 60 845E9-gelonin Gelonin 50 83HB21-bryodin II Bryodin 50 82HB21-luffin Luffin 50 79B2/25-saporin Saporin 50 81HB21-�-sarcin �-sarcin 30 80RSFv Restrictocin 40 72Anti-TfR(Fv)-EDN EDN 500 70Anti-TfR(Fv)AngL2 Angiogenin 5,000 71Anti-TfR(Fv)-pancRNase PancRNase 100,000 73

Abbreviations: DT, diphtheria toxin; PE, Pseudomonas exo-toxin; TfR, transferrin receptor; EDN, eosinophil-derived neu-rotoxin; pancRNase, pancreatic ribonuclease; Ang, angiogenin

IMMUNOTOXIN THERAPY OF HEMATOLOGIC MALIGNANCIES 549

Page 6: Immunotoxin therapy of hematologic malignancies

enzymatic activity. The conjugation of ligand andtoxin has employed both chemical conjugationand genetic fusion. The former has been accom-plished with bifunctional reagents such as thethiolating compounds—3-(2-pyridyldithio)propri-onic acid N-hydroxysuccinimide ester (SPDP) or3-maleimidobenzoic acid N-hydroxysuccinimideester (MBS). Such chemical conjugates bind to avariety of lysines on the ligand and/or toxin yield-ing a heterogeneous production with at least mildto moderate effects on ligand and toxin func-tions.2,4-8,10-12,15,16,18-21,23-31,46-56,59-65,79-84,86,87 Moresuccessful immunotoxin design has employed ge-netic engineering where an amide bond with orwithout a linker peptide connects the ligand andtoxin. Such fusions have been more successful ispreserving receptor affinity and toxin domainfunctions.3,4,9,13,17,22,32-45,57,58,66-74,76-78,85

CLINICAL RESULTS WITHIMMUNOTOXINS

Among the several hundred hematologic cancerimmunotoxins discussed above, about two dozenhave been tested in phase I-III clinical trials. Ex-citingly, a number of these compounds haveshown significant antileukemia and antilymphoma

activity, and one (DAB389IL2 or ONTAK, LigandPharmaceuticals, San Diego, CA) has been ap-proved by the US FDA for therapy of cutaneousT-cell lymphoma. Table 4 lists the published stud-ies of immunotoxins in hematologic malignancypatients. The table lists the compounds, the dis-eases for which they were tested, their responserates, and their toxicities. Direct comparisons aredifficult due to the different conjugate construc-tions, the different patient populations, and thedifferent routes, doses, and schedules of drug usedin the various trials. Nevertheless, we will addressthe efficacy, pharmacokinetics, immune responses,and toxicities for each agent and try to presentcommon principles.

Efficacy

Immunotoxins with the highest response rate inclinical studies have been active in tissue culturein the picomolar range and have been fully recom-binant molecules.

BL22 consists of a disulfide-stabilized anti-CD22sFv fused to PE38. When given at doses of 3 to 50�g/kg intravenously over 30 minutes every otherday for three doses to patients with purine analog-resistant hairy cell leukemia (HCL), there were 13

Table 4. Results of Clinical Studies of Hematologic Neoplasm Immunotoxins

Immunotoxin Disease Response Rate Toxicities References

RFB4(dsFv)-PE38 (BL22) Hairy cell leukemia 13/16 HUS 88Anti–Tac(Fv)-PE38 (LMB-2) Lymphoma 8/35 Transaminasemia 89DAB389IL2 (ONTAK) CTCL 33/91 VLS, transaminasemia 90, 93B43-PAP B-cell ALL 5/17 VLS 95IgRFB4-dg. ricin A Lymphomas 11/45 VLS 96-98FabRFB4-dg ricin A Lymphomas 5/14 VLS 101BerH2-saporin Hodgkin’s disease 3/4 None 102DAB486IL2 Lymphomas 12/109 Transaminasemia, HUS, renal insufficiency 103IgG-HD37-dg ricin A Lymphomas 5/54 VLS, HUS, acrocyanosis 104, 1053A1e-dg ricin A T-cell leukemias 2/11 VLS 106DT388GMCSF AML 4/37 Transaminasemia, liver failure 100Anti–B4-blocked ricin Lymphomas 8/75 VLS, transaminasemia, akinetic mutism 113-115T101-ricin A CLL 0/5 None 107H65-ricin A CLL, CTCL 2/10 CLL; 4/14 CTCL VLS 108, 109SPV-T3a-dg ricin A/WT-1-dg

ricin A GVHD 2/4 None 99Anti–Tac-PE ATL 0/4 Liver failure 110IgRFT5-dg ricin A Hodgkin’s disease 2/15 VLS 111K1-4-dg ricin A Hodgkin’s disease 1/15 VLS 112

Abbreviations: HUS, hemolytic-uremic syndrome; VLS, vascular leak syndrome; dg, deglycosylated; PAP, pokeweed antiviral protein; ALL, acutelymphoblastic leukemia; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; CTCL, cutaneous T cell lymphoma; GVHD,graft-versus-host disease; PE, Pseudomonas exotoxin; ATL, adult T-cell leukemia; DT, diphtheria toxin; DAB, diphtheria toxin A and B chain.

550 FRANKEL ET AL

Page 7: Immunotoxin therapy of hematologic malignancies

of responses among 16 patients, including 11 com-plete remissions.88 The three nonresponders eitherreceived a low dose or had pre-existing toxin-neutralizing antibodies. The remissions were dura-ble with only three of 11 responders relapsing after10 to 23 months. Re-treatment again producedcomplete remissions. Thus, BL22 immunotoxinappears to be the current best salvage treatmentfor relapsed HCL patients.

LMB2 is composed of an anti-CD25 sFv fused toPE38. LMB2 has been administered to 35 patientswith lymphomas at doses of 2 to 63 �g/kg intra-venously over 30 minutes four times per day for 3days.89 A complete remission was observed inHCL, which was ongoing at 20 months. Therewere seven partial remissions distributed as fol-lows: three patients with HCL, one with cutane-ous T-cell lymphoma (CTCL), one with chroniclymphocytic leukemia (CLL), one with Hodgkin’sdisease (HD), and one with adult T-cell leukemia(ATL). Responders received at least 60 �g/kg totaldose of LMB2 per cycle. The duration of remis-sions have not been determined but exceeds 20and 6 months for two patients and 1 month for theremaining patients. Responding patients hadclearance of circulating malignant cells, improve-ment in skin lesions, and regression of lymphnodes and splenomegaly.

ONTAK is a fusion protein composed of thecatalytic and translocation domains of DT fused tohuman IL-2. Among 71 CTCL patients with stageIB to IVA disease refractory to other therapies andtreated with 9 or 18 �g/kg/d ONTAK for 5 daysevery 3 weeks, there were 21 remissions, includingseven complete remissions.90 The median durationof remission was 7 months. When combined withdexamethasone 8 mg/d, responses were seen in 12of 20 CTCL patients.91 ONTAK also produced apartial remission in a patient with peripheral T-cell lymphoma (PTCL).92 In the phase I study,ONTAK produced a durable complete remission(ongoing after 5 years) in a patient with trans-plant-refractory large cell lymphoma.93 Two pa-tients with low-grade lymphoma obtained partialremissions. Based on the above results, in 1999 theFDA made ONTAK the first approved immuno-toxin.94

B43-PAP consists of an anti-CD19 monoclonalantibody chemically conjugated to PAP. Among17 children with relapsed B-cell acute lymphocyticleukemia (ALL), there were four complete remis-

sions and one partial remission to five daily intra-venous infusions of 0.5 to 1,250 �g/kg B43-PAP.95

IgRFB4-dgA is composed of an anti-CD22 an-tibody coupled to deglycosylated ricin A chain.When given either by a 192-hour continuous in-fusion or by four infusions over 4 hours each tochemoresistant B-cell non-Hodgkin’s lymphoma(NHL) patients and one post-kidney transplantlymphoma, there were 11 remissions among 54treated patients.96-98 These included two completeremissions lasting 2 and 32� months and ninepartial remissions lasting 2, 2, 3, 3, 3, 5, 6, 8, and14 months.

A combination of anti-CD3 (SPV-T3a-dgA)and anti-CD7 (WT1-dgA) immunotoxins wasgiven as three or four infusions of 2 or 4 mg/m2 ofa 1:1 combination of the two conjugates to foursteroid-refractory patients with graft-versus-hostdisease (GVHD) and two nearly complete remis-sions were observed.99 One patient had a resolu-tion of grade 4 skin GVHD. The other patient hada resolution of garde 2 intestinal GVHD.

DT388GMCSF consists of the catalytic andtranslocation domains of DT fused to human GM-CSF. A 1- to 5-�g/kg quantity was administeredintravenously over 15 minutes daily for 5 days to37 patients with relapsed or refractory AML.100

One patient had a complete remission with recov-ery of normal hematopoeisis lasting 1 year. After ayear, she had falling blood counts and increasedmarrow blasts. She was again treated and showedclearance of marrow blasts on day 12, but devel-oped sepsis and died on day 21. Three other pa-tients had partial remissions lasting months withclearance of marrow blasts on day 30, but did notachieve recovery of normal hematopoiesis. In onecase, the patient received a cord blood transplantafter achievement of blast cytoreduction.

Responses have been observed with other im-munotoxins as noted in Table 4, including Fab-�RFB4-dg ricin A, BerH2-saporin, DAB486IL2,IgG-HD37-dg ricin A, anti–B4-blocked ricin,H65-ricin A, IgRFT5-dg ricin A, and Ki-4-dg ricinA, but these molecules have not undergone furtherdevelopment.

Pharmacokinetics and Tissue Distribution

Larger molecules have longer half-lives in thecirculation and poorer tissue penetration. Thehalf-life of monoclonal antibody conjugates withRTA, ricin, PAP, and PE ranged from 9 to 24hours.95,98,102,106 The molecular weights were all

IMMUNOTOXIN THERAPY OF HEMATOLOGIC MALIGNANCIES 551

Page 8: Immunotoxin therapy of hematologic malignancies

around 200,000 Mr. There was likely little clear-ance of these molecules via renal glomeruli, andpenetration into extravascular sites such as nodes,marrow, or skin nodules was poor.107,115 In con-trast, the smaller recombinant immunotoxins(60,000 to 70,000 Mr) have had shorter half-livesof 1 to 5 hours88,89,93,100,103 and should have betterpenetration into extravascular sites of disease.However, there are no reports to date regardingthe saturation of extravascular tumor deposits inclinical trials with recombinant immunotoxins.The shorter circulation times and increased vas-cular permeability of the smaller recombinant im-munotoxins may contribute to their greater anti-tumor efficacy and reduced endothelial toxicity(see below).

Immune Responses

Because most individuals in the United Statesare immunized with diphtheria toxoid as children,close of half of adults have anti-DT antibody titerspretreatment with DT conjugates.116 Further, be-tween 10% and 20% of patients have had previousPseudomonas infections (often subclinical) yield-ing pretreatment anti-PE antibody titers.117 Pa-tients exposed to castor oil may show anti-ricinantibodies. These circulating antibodies reducethe half-life and AUC for immunotoxins and, insome cases, neutralize cell cytotoxicity.90,100,118

Even if patients lack pretreatment immunity, afteradministration of the foreign protein, most pa-tients develop anti-immunotoxin antibodies. Theexceptions include CLL patients and some pa-tients with severe immunosuppression.

Toxicities

There are two general classes of side effects dueto immunotoxins. In some instances, the targetedtoxin receptor/antigen is present on normal tis-sues. This can lead to significant toxicities.DT388GMCSF reacts with normal liver macro-phages—Kupffer cells. Subsequent cytokine re-lease triggers dose-limiting hepatocyte damage.100

Toxicities that are independent of ligand havebeen observed in almost every immunotoxin clin-ical study. The similarity of the toxicities observedwith different immunotoxin trials suggests that thelesions are due to the toxin moieties. The fiveclasses of side effects are (1) transient constitu-tional symptoms with fever, chills, nausea, vomit-ing, myalgias, arthralgias, asthenia, and hypoten-sion; (2) transient hepatotoxicity with elevated

transaminases and, rarely, other liver enzymes; (3)transient vascular leak syndrome (VLS) consistingof edema, hypoalbuminemia, weight gain, and,rarely, dyspnea and aphasia; (4) assorted syn-dromes including rhabdomyolysis, hemolytic ure-mic syndrome, and renal insufficiency with pro-teinuria and renal tubular acidosis; and (5) allergicreactions. The toxicities may be due to binding ofthe toxin moieties to normal tissues. Reaction ofimmunotoxins with macrophages, lymphocytes, orendothelium may lead to cytokine release withconsequent constitutional symptoms and elevatedcirculating cytokines. Elevations of IL-6 andTNF-� have been reported in some but not allcases.88,96,98,100 Where studied, corticosteroids orinfliximab � rofecoxib appeared to dampen oreliminate this side effect.88,91 The hepatotoxicitymay be directly due to immunotoxin binding af-fected by the pI of the ligand in the conjugate orsecondary release of cytokine from macrophagesmay damage the liver.100,119,120 In most cases, im-pairments of hepatic function such as VII produc-tion or bilirubin clearance were not observed andthe liver injury was reversible. VLS is a syndromeobserved with IL-2, monoclonal antibodies, somechemotherapy drugs such as taxotere, and manyimmunotoxins. The onset of the syndrome is usu-ally 4 to 6 days after initiating therapy and it lasts4 to 10 days. “Leaky” capillaries throughout thebody produce low serum albumin, edema, weightgain, dyspnea, aphasia, rhabdomyolysis, and pro-teinuria. The abnormal vascular endothelium maybe intoxicated by the immunotoxin directly ormay be “activated” by elevated circulating cyto-kines. Evidence from tissue culture and animalmodels has been reported for a direct binding siteof toxins on vascular endothelial cells and for anindirect toxic effect of immunotoxin-triggered in-flammatory cytokines on endothelial cells.121,122

There do not appear to be effective methods ofprophylaxis, although paradoxically active hydra-tion appears to reduce the incidence and severityof VLS. Further, smaller recombinant immunotox-ins appear to have a lower incidence, possibly dueto their shorter circulating half-life. Excluding pa-tients with prior radiotherapy may also lower theincidence or severity of the VLS syndrome.86 Therare cases of hemolytic-uremic syndrome (HUS)and rhabdomyolysis may be patient-specific typesof vascular injury or due to ligand (particularlyanti-CD22 antibodies). Allergic or anaphylactoidreactions occur very rarely, are IgE-mediated, and

552 FRANKEL ET AL

Page 9: Immunotoxin therapy of hematologic malignancies

are treated similar to allergic reactions to otherforeign proteins such as L-asparaginase. Fluids, car-diac monitoring, oxygen, corticosteroids, diphen-hydramine, and, rarely, epinephrine can be given.Patients with anaphylactoid reactions should notbe re-treated with the same immunotoxin.

CURRENT IMMUNOTOXIN CLINICALTRIALS IN HEMATOLOGIC

MALIGNANCIES

Current and near future applications of immu-notoxins in hematologic malignancies are focusedon expanding the use of some of the active, estab-lished agents, and testing novel compounds. Thesestudies and the sites for the studies are listed inTable 5.

Further protocols with DAB389IL2 (ONTAK)include testing in CD25� CTCL, B-cell NHL,PTCL,92 GVHD,123 and CLL.124 Combinationstudies with dexamethasone, bexarotene (an RXR-agonist), or tributyrin are being tested in CTCL,NHL, and CLL.91 Bexarotene and butyrates up-regulate IL-2R subunits on CTCL and CLL cellsand sensitize the malignant cells to killing byDAB389IL2.125,126 RFB4(Fv)-PE38 (BL22) is beingtested in HCL, CD22� HCL B-cell NHL, and

CD22� B-cell ALL. Anti-Tac(Fv)-PE38 (LMB-2)is being tested in patients with CLL and CTCL.DT388GMCSF is being tested in adults with re-lapsed AML in the absence of corticosteroids.SPV-T3a-dgA plus WT1-dgA is continuing phaseII evaluation for therapy of steroid-refractoryGVHD.

A number of novel immunotoxins have startedor will soon start clinical trials. HuM195-recom-binant gelonin has been given to 20 AML patientsto date and dose escalation is proceeding.DT388IL3 has been aseptically vialed, is undergo-ing monkey toxicology testing, and will enter clin-ical trials in the spring. BU12-saporin is beinggiven to children and adults with B-cell ALL andNHL, respectively. OKT10-saporin is being testedin patients with advanced multiple myeloma.A-dmDT390-bisFv is undergoing production andwill enter phase I study in patients with refractoryaplastic anemia and T-cell malignancies.

These studies should help define the role forimmunotoxins in the care for patients with hema-tologic malignancies. The last two decades havesuggested potential clinical applications for thesechimeric proteins. The next few years should seeimproved success of these agents as improvements

Table 5. Ongoing and Planned Immunotoxin Studies in Patients With Hematologic Neoplasms

Agent Target Receptor Disease Contact (telephone)

DAB389IL2 (ONTAK) IL-2R CD25� CTCL Foss (617-636-5558)NHL Dang (713-792-2860)GVHD Ho (617-632-5938)MDS Rosmarin (401-793-4648)

�bexarotene (Targretin) CTCL, NHL, CLL Foss (617-636-5558)�bexarotene (Targretin) CLL Frankel (336-716-3313)

RFB4dsFv-PE38(BL22) CD22 HCL Kreitman (301-402-5633)CLL, NHL Kreitman (301-402-5633)ALL Wayne (301-435-2257)

Anti-Tac(Fv)-PE38 (LMB2) CD25 CLL Kreitman (301-402-5633)CTCL Kreitman (301-402-5633)

DT388GMCSF GM-CSFR AML Sweetenham (303-315-5608)DT388IL3 IL-3R AML Rizzieri (919-684-8111)A-dmDT390bisFv CD3� AA, T-NHL Frankel (336-716-3313)HuM195-rgelonin CD33 AML Talpaz (713-792-3525)BU12-saporin CD19 B-cell ALL Flavell (44-1703796947)OKT10-saporin CD38 Myeloma Flavell (44-1703796947)SPV-T3a-dgA/WT1-dgA CD3/CD7 GVHD De Witte (31-2-4361476)

Abbreviations: AA, aplastic anemia; NHL, non-Hodgkin’s lymphoma; HD, Hodgkin’s disease; GVHD, graft-versus-host disease; ALL, acutelymphoblastic leukemia; AML, acute myeloid leukemia; HCL, hairy cell leukemia; MDS, myelodysplastic syndrome; CTCL, cutaneous T-celllymphoma.

IMMUNOTOXIN THERAPY OF HEMATOLOGIC MALIGNANCIES 553

Page 10: Immunotoxin therapy of hematologic malignancies

are made in protein design to reduce normal tissuetoxicities and the new molecules are matched withthe molecular profile of particular patients to en-hance response rates.

REFERENCES

1. Tabbara IA, Zimmerman K, Morgan C, et al: Allogeneichematopoietic stem cell transplantation: Complications andresults. Arch Intern Med 162:1558-1566, 2002

2. Tazzari PL, Bolognesi A, De Totero D, et al: Immuno-toxins containing saporin linked to different CD2 monoclonalantibodies: In vitro evaluation. Br J Haematol 86:97-105, 1994

3. Woo JH, Liu YY, Mathias A, et al: Gene optimization isnecessary to express a bivalent anti-human anti-T cell immu-notoxin in Pichia pastoris. Protein Expr Purif 25:270-282, 2002

4. Barbieri L, Bolognesi A, Dinota A, et al: Selectivekilling of CD4� and CD8� cells with immunotoxins contain-ing saporin. Scand J Immunol 30:369-372, 1989

5. Better M, Bernhard SL, Fishwild DM, et al: Geloninanalogs with engineered cysteine residues form antibody im-munoconjugates with unique properties. J Biol Chem 269:9644-9450, 1994

6. Press OW, Martin PJ, Thorpe PE, et al: Ricin A-chaincontaining immunotoxins directed against different epitopeson the CD2 molecule differ in their ability to kill normal andmalignant T cells. J Immunol 141:4410-4417, 1988

7. Derocq JM, Casellas P, Laurent G, et al: Comparison ofthe cytotoxic potency of T101 Fab, F(ab’)2 and whole IgGimmunotoxins. J Immunol 141:2837-2843, 1988

8. Collinson AR, Lambert JM, Liu Y, et al: Anti-CD6-blocked ricin: An anti-pan T-cell immunotoxin. Int J Immu-nopharmacol 16:37-49, 1994

9. Pauza ME, Doumbia SO, Pennell CA: Construction andcharacterization of human CD7-specific single-chain Fv immu-notoxins. J Immunol 158:3259-3269, 1997

10. Preijers FWMB, Tax WJM, De Witte T, et al: Relation-ship between internalization and cytotoxicity of ricin A-chainimmunotoxins. Br J Haematol 70:289-294, 1988

11. Luo Y, Seon BK: Marked difference in the in vivoantitumor efficacy between two immunotoxins targeted to dif-ferent epitopes of common acute lymphoblastic leukemia an-tigen (CD10). J Immunol 145:1974-1982, 1990

12. Flavell DJ, Flavell SU, Boehm DA, et al: Preclinicalstudies with the anti-CD19-saporin immunotoxin BU12-sa-porin for the treatment of human B-cell tumours. Br J Cancer72:1373-1379, 1995

13. Salvatore G, Beers R, Margulies I, et al: Improved cyto-toxic activity toward cell lines and fresh leukemia cells of amutant anti-CD22 immunotoxin obtained by antibody phagedisplay. Clin Cancer Res 8:995-1002, 2002

14. Onda M, Kreitman RJ, Vasmatzis G, et al: Reduction ofthe nonspecific animal toxicity of anti-Tac(Fv)-PE38 by mu-tations in the framework regions of the Fv which lower theisoelectric point. J Immunol 163:6072-6077, 1999

15. Duke-Cohan JS, Morimoto C, Schlossman SF: Target-ing of an activated T-cell subset using a bispecific antibody-toxin conjugate directed against CD4 and CD26. Blood 82:2224-2234, 1993

16. Duke-Cohan JS, Morimoto C, Schlossman SF: Deple-tion of the helper/inducer (memory) T cell subset using a

bispecific antibody-toxin conjugate directed against CD4 andCD29. Transplantation 56:1188-1196, 1993

17. Nagata S, Onda M, Numata Y, et al: Novel anti-CD30recombinant immunotoxins containing disulfide-stabilized Fvfragments. Clin Cancer Res 8:2345-2355, 2002

18. Pagliaro LC, Liu B, Munker R, et al: Humanized M195monoclonal antibody conjugated to recombinant gelonin: Ananti-CD33 immunotoxin with antileukemic activity. ClinCancer Res 4:1971-1976, 1998

19. Bonardi MA, French RR, Amlot P, et al: Delivery ofsaporin to human B-cell lymphoma using bispecific antibody:targeting via CD22 but not CD19, CD37 or immunoglobulinresults in efficient killing. Cancer Res 53:3015-3121, 1993

20. Engert A, Brown A, Thorpe P: Resistance of myeloidleukaemia cell lines to ricin A-chain immunotoxins. Leuk Res15:1079-1086, 1991

21. Flavell DJ, Boehm DA, Noss A, et al: Therapy of humanT-cell acute lymphoblastic leukaemia with a combination ofanti-CD7 and anti-CD38-saporin immunotoxins is signifi-cantly better than therapy with each individual immunotoxin.Br J Cancer 84:571-578, 2001

22. Francisco JA, Gawlak SL, Siegall CB: Construction,expression and characterization of BD1-G28-5 sFv, a single-chain anti-CD40 immunotoxin containing the ribosome-inac-tivating protein bryodin 1. J Biol Chem 272:24165-24169,1997

23. Shumakov VI, Maisjuk IG, Agapov II, et al: In vitroefficacy of conjugates of anti-CD45 monoclonal antibodieswith plant toxin A-chains. Transplant Proc 30:971-973, 1998

24. McCoig C, Van Dyke G, Chou C-S, et al: An anti-CD45RO immunotoxin eliminates T cells latently infectedwith HIV-1 in vitro. Proc Natl Acad Sci USA 96:11482-11485, 1999

25. Zangemeister-Wittke U, Lehmann H-P, Waibel R, et al:Action of a CD24-specific deglycosylated ricin-A-chain immu-notoxin in conventional and novel models of small-cell-lung-cancer xenograft. Int J Cancer 53:521-528, 1993

26. Tazzari PL, Barbieri L, Gobbi M, et al: An immunotoxincontaining a rat IgM monoclonal antibody (Campath 1) andsaporin 6: Effect on T lymphocytes and hemopoietic cells.Cancer Immunol Immunother 26:231-236, 1988

27. Huang YW, Burrows FJ, Vitetta ES: Cytotoxicity of anovel anti-ICAM-1 immunotoxin on human myeloma celllines. Hybridoma 12:661-675, 1993

28. Zangemeister-Wittke U, Collinson AR, Frosch B, et al:Immunotoxins recognizing a new epitope on the neural celladhesion molecule have potent cytotoxic effects against smallcell lung cancer. Br J Cancer 69:32-39, 1994

29. Zhong RK, van de Winkel JG, Thepen T, et al: Cyto-toxicity of anti-CD64-ricin A chain immunotoxin against hu-man acute myeloid leukemia cells in vitro and in SCID mice.J Hematother Stem Cell Res 10:95-105, 2001

30. Myers DE, Uckun FM: An anti-CD72 immunotoxinagainst therapy-refractory B-lineage acute lymphoblastic leuke-mia. Leuk Lymphoma 18:119-122, 1995

31. Bolognesi A, Polito L, Tazzari PL, et al: In vitro anti-tumour activity of anti-CD80 and anti-CD86 immunotoxinscontaining type 1 ribosome-inactivating proteins. Br J Haema-tol 110:351-361, 2000

32. Kreitman RJ, Schneider WP, Queen C, et al: Mik-beta1(Fv)-PE40, a recombinant immunotoxin cytotoxic to-

554 FRANKEL ET AL

Page 11: Immunotoxin therapy of hematologic malignancies

ward cells bearing the beta-chain of the IL-2 receptor. J Im-munol 149:2810-2815, 1992

33. Re GG, Waters C, Poisson L, et al: Interleukin 2 (IL-2)receptor expression and sensitivity to diphtheria fusion toxinDAB389IL-2 in cultured hematopoietic cells. Cancer Res 56:2590-2595, 1996

34. Frankel AE, Ramage J, Kiser M, et al: Characterizationof diphtheria fusion proteins targeted to the human interleu-kin-3 receptor. Protein Engineer 13:575-581, 2000

35. Cai J, Zheng T, Murphy J, et al: IL-4R expression inAIDS-KS cells and response to rhIL-4 and IL-4 toxin (DAB389-IL-4). Invest New Drugs 15:279-287, 1997

36. Siegall CB, Kreitman RJ, FitzGerald DJ, et al: Antitu-mor effects of interleukin-6-Pseudomonas exotoxin chimericmolecules against the human hepatocellular carcinoma, PLC/PRF/5 in mice. Cancer Res 51:2831-2836, 1991

37. Sweeney EB, Foss FM, Murphy JR, et al: Interleukin 7(IL-7) receptor-specific killing by DAB389IL-7: A novel agentfor the elimination of IL-7 receptor positive cells. BioconjChem 9:201-207, 1998

38. Klimka A, Barth S, Drillich S, et al: A deletion mutantof Pseudomonas exotoxin-A fused to recombinant human in-terleukin-9 (rhIL-9-ETA�) shows specific cytotoxicity againstIL-9-receptor-expressing cell lines. Cytokines Mol Ther 2:139-146, 1996

39. Oshima Y, Puri RK: Suppression of an IL-13 autocrinegrowth loop in a human Hodgkin/Reed-Sternberg tumor cellline by a novel IL-13 antagonist. Cell Immunol 211:37-42,2001

40. vanderSpek JC, Sutherland J, Sampson E, et al: Geneticconstruction and characterization of the diphtheria toxin-re-lated interleukin-15 fusion protein DAB389sIL15. Protein En-gineer 8:1317-1321, 1995

41. Tojo A, Oshima Y: Targeted toxin therapy in the treat-ment of leukemia. Jpn J Clin Hematol 6:578-581, 1995

42. Frankel AE, Ramage J, Latimer A, et al: High-levelexpression and purification of the recombinant diphtheria fu-sion toxin DTGM for phase I clinical trials. Protein Expr Purif16:190-201, 1999

43. Ramage JG, Vallera DA, Black JH, et al: The diphtheriatoxin/urokinase fusion protein DTAT is selectively toxic toCD87 expressing leukemic cells. Leuk Res (in press)

44. Bruhl H, Cihak J, Stangassinger M, et al: Depletion ofCCR5-expressing cells with bispecific antibodies and chemo-kine toxins: A new strategy in the treatment of chronic in-flammatory diseases and HIV. J Immunol 166:2420-2426, 2001

45. Fisher CE, Sutherland JA, Krause JE, et al: Geneticconstruction and properties of a diphtheria toxin-related sub-stance P fusion protein: In vitro destruction of cells bearingsubstance P receptors. Proc Natl Acad Sci USA 93:7341-7345,1996

46. Chiron M, Jaffrezou JP, Carayon P, et al: Induction andincrease of HLA-DR antigen expression by immune interferonon ML-3 cell line enhances the anti-HLA-DR immunotoxinactivity. Clin Exp Immunol 82:214-220, 1990

47. Wiels J, Junqua S, Dujardin P, et al: Properties of im-munotoxins against a glycolipid antigen associated with Bur-kitt’s lymphoma. Cancer Res 44:129-133, 1984

48. Faguet GB, Agee JF: Four ricin chain A-based immuno-toxins directed against the common chronic lymphocytic leu-kemia antigen: in vitro characterization. Blood 82:536-543,1993

49. Tonevitsky AG, Agapov II, Ershova GV, et al: Cyto-toxic effect of ricin A-chain conjugates containing monoclonalantibodies against human erythroid cells. Intl J Immunopharm15:229-235, 1993

50. Bregni M, Lappi DA, Siena S, et al: Activity of amonoclonal antibody-saporin-6 conjugate against B-lymphomacells. J Natl Cancer Inst 80:511-517, 1988

51. Avila AD, de Acosta M, Lage A: A new immunotoxinbuilt by linking a hemolytic toxin to a monoclonal antibodyspecific for immature T lymphocytes. Int J Cancer 42:568-571,1988

52. Barbieri L, Dinota A, Gobbi M, et al: Immunotoxinscontaining saporin 6 and monoclonal antibodies recognizingplasma cell-associated antigens: Effects on target cells and onnormal myeloid precursors (CFU-GM). Eur J Haematol 42:238-245, 1989

53. Yoshida M, Rybak RJ, Choi Y, et al: Development ofa severe combined immunodeficiency (SCID) mouse modelconsisting of highly disseminated human B-cell leukemia/lymphoma, cure of the tumors by systemic administration ofimmunotoxin, and development/application of a clonotype-specific polymerase chain reaction-based assay. Cancer Res57:678-685, 1997

54. Tecce R, Fraioli R, De Frabritiis P, Sandrelli A, et al:Production and characterization of two immunotoxins specificfor M5b ANLL leukaemia. Int J Cancer 49:310-316, 1991

55. Harris P, Reed E, King DW, et al: In vitro studies of theeffect of Mab NDA 4 linked to toxin on the proliferation of ahuman EBV-transformed lymphoblastoid B cell line and ofgibbon MLA leukemia cell line. Cell Immunol 134:85-95, 1991

56. Poccia F, Piselli P, Di Cesare S, et al: Recognition andkilling of tumour cells expressing heat shock protein 65kD withimmunotoxins containing saporin. Br J Cancer 66:427-432,1992

57. Chovnick A, Schneider WP, Tso JY, et al: A recombi-nant membrane-activing immunotoxin. Cancer Res 51:465-467, 1991

58. Choo ABH, Dunn RD, Broady KW, et al: Soluble ex-pression of a functional recombinant cytolytic immunotoxin ininsect cells. Protein Expr Purif 24:338-347, 2002

59. Gasanov SE, Rael ED, Gasnov NE, et al: In vitro eval-uation of Pyrularia thionin-anti-CD5 immunotoxin. CancerImmunol Immunother 41:122-128, 1995

60. Hinman CL, Tang HP: A membrane-lytic immunocon-jugate selective for human tumor T-lymphocytes. Intl J Immu-nopharm 20:467-478, 1998

61. Tonevitsky AG, Agapov II, Shamshiev AT, et al: Im-munotoxins containing A-chain of mistletoe lectin I are moreactive than immunotoxins with ricin A-chain. FEBS Lett 392:166-168, 1996

62. Dinota A, Barbieri L, Gobbi M, et al: An immunotoxincontaining momordin suitable for bone marrow purging inmultiple myeloma patients. Br J Cancer 60:315-319, 1989

63. Bolognesi A, Tazzari PL, Legname G, et al: Anti-CD30immunotoxins with native and recombinant dianthin 30. Can-cer Immunol Immunother 40:109-114, 1995

64. Bolognesi A, Tazzari PL, Tassi C, et al: A comparison ofanti-lymphocyte immunotoxins containing different ribosome-inactivating proteins and antibodies. Clin Exp Immunol 89:341-346, 1992

65. Better M, Bernhard SL, Lei SP, et al: Activity of recom-

IMMUNOTOXIN THERAPY OF HEMATOLOGIC MALIGNANCIES 555

Page 12: Immunotoxin therapy of hematologic malignancies

binant mitogillin and mitogillin immunoconjugates. J BiolChem 267:16712-16718, 1992

66. Newton DL, Hansen HJ, Mikulski SM, et al: Potent andspecific antitumor effects of an anti-CD22-targeted cytotoxicribonuclease: potential for the treatment of non-Hodgkin’slymphoma. Blood 97:528-535, 2001

67. Newton DL, Ilercil O, Laske DW, et al: Cytotoxicribonuclease chimeras. J Biol Chem 267:19572-15978, 1992

68. Aqeilan R, Yarkoni S, Loberboum-Galski H: Interleukin2-Bax: A novel prototype of human chimeric proteins fortargeted therapy. FEBS Lett 457:271-276, 1999

69. Psarras K, Ueda M, Tanabe M, et al: Targeting activatedlymphocytes with an entirely human immunotoxin analogue:Human pancreatic RNase 1-human IL-2 fusion. Cytokine 12:786-790, 2000

70. Newton DL, Nicholls PJ, Rybak SM, et al: Expressionand characterization of recombinant eosinophil-derived neuro-toxin and eosinophil-derived neurotoxin-anti-transferrin re-ceptor sFv. J Biol Chem 269:26739-26745, 1994

71. Newton DL, Xue Y, Olson KA, et al: Angiogenin single-chain immunofusions: Influence of peptide linkers and spacersbetween fusion protein domains. Biochemistry 35:545-553,1996

72. Goyal A, Batra JK: Inclusion of a furin-sensitive spacerenhances the cytotoxicity of ribotoxin restrictocin containingrecombinant single-chain immunotoxins. Biochem J 345:247-254, 2000

73. Rybak S, Saxena SK, Ackerman EJ, et al: Cytotoxicpotential of ribonuclease and ribonuclease hybrid proteins.J Biol Chem 266:21202-21207, 1991

74. Burbage C, Tagge EP, Harris B, et al: Ricin fusion toxintargeted to the human granulocyte-macrophage colony stimu-lating factor receptor is selectively toxic to acute myeloidleukemia cells. Leuk Res 21:681-690, 1997

75. Baluna R, Rizo J, Gordon BE, et al: Evidence for astructural motif in toxins and interleukin-2 that may be respon-sible for binding to endothelial cells and initiating vascular leaksyndrome. Proc Natl Acad Sci USA 96:3957-3962, 1999

76. Vallera DA, Kuroki DW, Panoskaltsis-Mortari A,et al: Molecular modification of a recombinant anti-CD3�-directed immunotoxin by inducing terminal cysteine bridgingenhances anti-GVHD efficacy and reduces organ toxicity in alethal murine model. Blood 96:1157-1165, 2000

77. Liu S, Bugge TH, Leppla SH: Targeting of tumor cells bycell surface urokinase plasminogen activator-dependent an-thrax toxin. J Biol Chem 276:17976-17984, 2001

78. Liu S, Netzel-Arnett S, Birkedal-Hansen H, et al: Tu-mor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxin. Cancer Res 60:6061-6067, 2000

79. Ramakrishnan S, Enghlid JJ, Bryant HL, et al: Charac-terization of a translation inhibitory protein from Luffa aegyp-tiaca. Biochem Biophys Res Commun 160:509-516, 1989

80. Rahtore D, Nayak S, Batra JK: Overproduction of fungalribotoxin �-sarcin in Escherichia coli: Generation of an activeimmunotoxin. Gene 190:31-35, 1997

81. Cazzola M, Bergamaschi G, Dezza L, et al: Cytotoxicactivity of an anti-transferrin receptor immunotoxin on normaland leukemic human hematopoietic progenitors. Cancer Res51:536-541, 1991

82. Ebert RF, Spryn LA: Immunotoxin construction with aribosome-inactivating protein from barley. Bioconj Chem1:331-336, 1990

83. Scott CF, Goldmacher VS, Lambert JM, et al: An im-munotoxin composed of a monoclonal antitransferrin receptorantibody linked by a disulfide bond to the ribosome-inactivat-ing protein gelonin: Potent in vitro and in vivo effects againsthuman tumors. J Natl Cancer Inst 79:1163-1172, 1987

84. Bjorn M, Ring D, Frankel AE: Evaluation of monoclonalantibodies for the development of breast cancer immunotoxins.Cancer Res 45:1214-1221, 1985

85. Kreitman RJ, Pastan I: Recombinant toxins containinghuman granulocyte-macrophage colony-stimulating factor andeither Pseudomonas exotoxin or diphtheria toxin kill gastroin-testinal cancer and leukemia cells. Blood 90:252-259, 1997

86. Schindler J, Sausville E, Messmann R, et al: The toxicityof deglycosylated ricin A chain-containing immunotoxins inpatients with non-Hodgkin’s lymphoma is exacerbated by priorradiotherapy: A retrospective analysis of patients in five clinicaltrials. Clin Cancer Res 7:255-258, 2001

87. Huang CA, Yamada K, Murphy MC, et al: In vivo T celldepletion in miniature swine using the swine CD3 immuno-toxin, pCD3-CRM9. Transplantation 68:855-860, 1999

88. Kreitman RJ, Wilson WH, Bergeron K, et al: Efficacy ofthe anti-CD22 recombinant immunotoxin BL22 in chemother-apy-resistant hairy-cell leukemia. N Engl J Med 345:241-247,2001

89. Kreitman RJ, Wilson WH, White JD, et al: Phase I trialof recombinant immunotoxin anti-Tac(Fv)-PE38 (LMB-2) inpatients with hematologic malignancies. J Clin Oncol 18:1622-1636, 2000

90. Olsen E, Duvic M, Frankel A, et al: Pivotal phase IIItrial of two dose levels of denileukin diftitox for the treatmentof cutaneous T-cell lymphoma. J Clin Oncol 19:376-388, 2001

91. Foss FM, Bacha P, Osann KE, et al: Biological correlatesof acute hypersensitivity events with DAB(389)IL-2 (denileu-kin diftitox, ONTAK) in cutaneous T-cell lymphoma: De-creased frequency and severity with steroid premedication. ClinLymphoma 1:298-302, 2001

92. Talpur R, Apisarnthanarax N, Ward S, et al: Treatmentof refractory peripheral T-cell lymphoma with denileukin difti-tox (ONTAK). Leuk Lymphoma 43:121-126, 2002

93. LeMaistre CF, Saleh MN, Kuzel TM, et al: Phase I trialof a ligand fusion-protein (DAB389IL-2) in lymphomas express-ing the receptor for interleukin-2. Blood 91:399-405, 1998

94. Piascik P: FDA approves fusion protein for treatment oflymphomaJ Am Pharm Assoc391999571572 UckunFMAnno-tation: Immunotoxins for the treatment of leukaemia. Br JHaematol 85:435-438, 1993

95. Uckun FM: Annotation: Immunotoxins for the treat-ment of leukaemia . Br J Haematol 85:435-438, 1993

96. Sausville EA, Headlee D, Stetler-Steven M, et al: Con-tinuous infusion of the anti-CD22 immunotoxin IgG-RFB4-SMPT-dgA in patients with B-cell lymphoma: A phase I study.Blood 85:3457-3465, 1995

97. Senderowicz AM, Vitetta E, Headlee D, et al: Completesustained response of a refractory, post-transplantation, largeB-cell lymphoma to an anti-CD22 immunotoxin. Ann InternMed 126:882-885, 1997

98. Amlot PL, Stone MJ, Cunningham D, et al: A phase Istudy of an anti-CD22-deglycosylated ricin A chain immuno-toxin in the treatment of B-cell lymphomas resistant to con-ventional therapy. Blood 82:2624-2633, 1993

99. Van Oosterhout YVJM, van Emst L, SchattenbergAVMB, et al: A combination of anti-CD3 and anti-CD7 ricin

556 FRANKEL ET AL

Page 13: Immunotoxin therapy of hematologic malignancies

A-immunotoxins for the in vivo treatment of acute graft versushost disease. Blood 95:3693-3701, 2000

100. Frankel AE, Powell BL, Hall PD, et al: Phase I trialof a novel diphtheria toxin/GM-CSF fusion protein(DT388GMCSF) for refractory or relapsed acute myeloid leu-kemia (AML). Clin Cancer Res 8:1004-1013, 2002

101. Vitetta ES, Stone M, Amlot P, et al: Phase I immuno-toxin trial in patients with B-cell lymphoma. Cancer Res51:4052-4058, 1991

102. Falini B, Bolognesi A, Flenghi L, et al: Response ofrefractory Hodgkin’s disease to monoclonal anti-CD30 immu-notoxin. Lancet 339:1195-1196, 1992

103. VanderSpek J, Cosenza L, Woodworth T, et al: Diph-theria toxin-related cytokine fusion proteins: Elongation factor2 as a target for the treatment of neoplastic disease. Mol CellBiochem 138:151-156, 1994

104. Stone MJ, Sausville EA, Fay JW, et al: A phase I studyof bolus versus continuous infusion of the anti-CD19 immuno-toxin, IgG-HD37-dgA, in patients with B-cell lymphoma.Blood 88:1188-1197, 1996

105. Messman RA, Vitetta ES, Headlee D, et al: A phase Istudy of combination therapy with immunotoxins IgG-HD37-deglycosylated ricin A chain (dgA) and IgG-RFB4-dgA (Com-botox) in patients with refractory CD19�, CD22� B cell lym-phoma. Clin Cancer Res 6:1302-1313, 2000

106. Frankel AE, Laver JH, Willingham MC, et al: Therapyof patients with T-cell lymphomas and leukemias using ananti-CD7 monoclonal antibody-ricin A chain immunotoxin.Leuk Lymphoma 26:287-298, 1997

107. Hertler AA, Schlossman DM, Borowitz MJ, et al: Ananti-CD5 immunotoxin for chronic lymphocytic leukemia: En-hancement of cytotoxicity with human serum albumin-mon-ensin. Int J Cancer 43:215-219, 1989

108. LeMaistre CF, Rosen S, Frankel A, et al: Phase I trialof H65-RTA immunoconjugate in patients with cutaneousT-cell lymphoma. Blood 78:1173-1182, 1991

109. LeMaistre CF, Deisseroth A, Foget B, et al: Phase I trialof H65-RTA in patients with chronic lymphocytic leukemia.Blood 76:295a, 1990 (suppl 1, abstr)

110. Waldman T, Pastan I, Gansow O, et al: The mul-tichain interleukin-2 receptor: A target for immunotherapy.Ann Intern Med 116:148-160, 1992

111. Engert A, Diehl V, Schnell R, et al: A phase I study ofan anti-CD25 ricin A-chain immunotoxin (RFT5-SMPT-dgA)in patients with refractory Hodgkin’s lymphoma. Blood 89:403-410, 1997

112. Schnell R, Staak O, Borchmann P, et al: A phase Istudy of an anti-CD30 ricin A-chain immunotoxin (Ki-4. dgA)in patients with refractory CD30� Hodgkin’s and non-Hodgkin’s lymphoma. Clin Cancer Res 8200217791786

113. Grossbard ML, Freedman AS, Ritz J, et al: Serotherapyof B-cell neoplasms with anti-B4-blocked ricin: A phase I trialof daily bolus infusion. Blood 79:576-585, 1992

114. Grossbard ML, Lambert JM, Goldmacher VS, et al:Anti-B4-blocked ricin: A phase I trial of 7-day continuousinfusion in patients with B-cell neoplasms. J Clin Oncol 11:726-737, 1993

115. Multani PS, O’Day S, Nadler LM, et al: Phase IIclinical trial of bolus infusion anti-B4 blocked ricin immuno-conjugate in patients with relapsed B-cell non-Hodgkin’s lym-phoma. Clin Cancer Res 4:2599-2604, 1998

116. Hall PD, Virella G, Beckham W, et al: Adult AMLpatients treated with DTGM diphtheria fusion toxin do nothave neutralizing antibodies before or after the first treatmentcourse. Clin Immunol 100:191-197, 2001

117. Rose DM, Pai LH, Pastan I: Identification of epitopeson a mutant form of Pseudomonas exotoxin using serum fromhumans treated with Pseudomonas exotoxin containing immu-notoxins. Eur J Immunol 27:1459-1468, 1997

118. Hertler AA, Spitler LE, Frankel AE: Humoral immuneresponse to a ricin A chain immunotoxin in patients withmetastatic melanoma. Cancer Drug Deliv 4:245-253, 1987

119. Onda M, Nagata S, Tsutsumi Y, et al: Lowering theisoelectric point of the Fv portion of recombinant immunotox-ins leads to decreased nonspecific animal toxicity without af-fecting antitumor activity. Cancer Res 61:5070-5077, 2001

120. Onda M, Willingham MC, Wang QC, et al: Inhibitionof TNF-alpha produced by Kupffer cells protects against thenonspecific liver toxicity of immunotoxin anti-Tac(Fv)-PE38,LMB-2. J Immunol 165:7150-7156, 2000

121. Baluna R, Coleman E, Jones C, et al: The effect of amonoclonal antibody coupled to ricin A chain-derived pep-tides on endothelial cells in vitro: insights into toxin-mediatedvascular damage. Exp Cell Res 258:417-424, 2000

122. Siegall CB, Liggitt D, Chace D, et al: Characterizationof vascular leak syndrome induced by the toxin component ofPseudomonas exotoxin-based immunotoxins and its potentialinhibition with nonsteroidal anti-inflammatory drugs. ClinCancer Res 3:339-345, 1997

123. Ho VT, Zahrieh D, Neuberg D, et al: Safety andefficacy of denileukin diftitox (ONTAK) in patients with ste-roid refractory graft-versus-host disease (GVHD) after alloge-neic hematopoietic stem cell transplantation (HSCT). Blood100:423a, 2002 (suppl 1, abstr)

124. Frankel AE, Fleming DR, Powell BL, et al: DAB389IL2(ONTAK) fusion protein therapy of chronic lymphocytic leu-kemia. Exp Opin Biol Ther (in press)

125. Gorgun G, Foss F: Immunomodulatory effects of RXRrexinoids: Modulation of high-affinity IL-2R expression en-hances susceptibility to denileukin diftitox. Blood 100:1399-1403, 2002

126. Shao RH, Tian X, Gorgun G, et al: Arginine butyrateincreases the cytotoxicity of DAB389IL-2 in leukemia andlymphoma cells by upregulation of IL-2R� gene. Leuk Res26:1077-1083, 2002

IMMUNOTOXIN THERAPY OF HEMATOLOGIC MALIGNANCIES 557