CCR FOCUS

Antibody Fusion Proteins: Anti-CD22 Recombinant

Robert J. Kreitman and Ira Pastan

Abstract Recombinant are fusion proteins that contain the cytotoxic portion of a protein toxin fused to the Fv portion of an . The Fv binds to an antigen on a target cell and brings the toxin into the cell interior, where it arrests protein synthesis and initiates the apoptotic cascade. Moxetumomab pasudotox, previously called HA22 or CAT-8015, is a recombinant immunotoxin composed of the Fv fragment of an anti- CD22 fused to a 38-kDa fragment of Pseudomonas exotoxin A, called PE38. Moxe- tumomab pasudotox is an improved, more active form of a predecessor recombinant immunotoxin, BL22 (also called CAT-3888), which produced complete remission in relapsed/refractory (HCL), butithada<20% response rate in chronic lymphocytic leukemia (CLL) and acute lymphoblastic leukemia (ALL), diseases in which the leukemic cells contain much lower numbers of CD22 target sites. Compared with BL22, moxetumomab pasudotox is up to 50-fold more active on lymphoma cell lines and leukemic cells from patients with CLL and HCL. A phase I trial was recently completed in HCL patients, who achieved response rates similar to those obtained with BL22 but without dose-limiting toxicity. In addition to further testing in HCL, moxetumomab pasudotox is being evaluated in phase I trials in patients with CLL, B-cell lymphomas, and childhood ALL. Moreover, protein engineering is being used to increase its activity, decrease nonspecific side effects, and remove B-cell epitopes. Clin Cancer Res; 17(20); 6398–405. 2011 AACR.

Introduction enables them to kill cells with relatively few target sites, and a unique mechanism of action that can bypass some Targeted toxins types of drug resistance. This review focuses on recom- Different approaches have been used and are under binant immunotoxins targeted to CD22, particularly development to target toxic molecules to cancer cells and moxetumomab pasudotox. kill them more efficiently than would be possible with monoclonal (mAb) alone. Radioimmunother- Protein toxins apy is the most established of these approaches, repre- Protein toxins produced by bacteria, fungi, and plants are sented by the approved drugs and extremely cytotoxic and can kill cells when only one or a few (1), and reviewed in this issue by Steiner molecules reach the cytosol (9–12). These agents act by and Neri (2). Exciting advances have been made with inhibiting protein synthesis and inducing apoptosis. The antibody-drug conjugates (3), particularly bacterial toxins Pseudomonas exotoxin A (PE) and diphtheria emtansine (4), SGN-35 (; ref. 5), toxin (DT) catalytically inactivate elongation factor 2 by SAR3419 (6), and calicheamicin conjugates (7). Cyto- ADP ribosylation (9, 10). Plant toxins inactivate ribosomes kines fused to antibodies are being used to attract by removing adenine4324 in 28s ribosomal RNA (11, 13). immune effector cells to cancer cells (8). Immunotoxins Unlike plant toxins, bacterial toxins are made as single- are a type of antibody-conjugate in which a powerful chain proteins, making them more amenable for construc- protein toxin instead of a low-molecular-weight drug is tion of recombinant chimeric proteins (14). Recombinant attached to an antibody or antibody fragment. Their immunotoxins are produced by replacing the binding possible advantages include a very high activity, which domain of the toxin with the Fv portion of an antibody that directs the toxin to a cancer cell. Target choice is very important to prevent killing of normal cells. The lineage- restricted differentiation B-cell antigen CD22 is an excellent Authors' Affiliation: Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, Maryland target because it is absent on normal tissues like liver and skin, and its lack of expression on B-cell precursors allows Corresponding Author: Robert J. Kreitman, Laboratory of Molecular þ Biology, National Cancer Institute, NIH, 37/5124b, 9000 Rockville Pike, normal CD22 B cells to be rapidly generated after immu- Bethesda, MD 20892. Phone: 301-496-6947; Fax: 800-864-7554; E-mail: notoxin therapy ceases. [email protected] CD22, a sialic acid–binding immunoglobulin-like lectin doi: 10.1158/1078-0432.CCR-11-0487 (siglec) that inhibits B-cell receptor calcium signaling, is 2011 American Association for Cancer Research. expressed on many B-cell malignancies, including hairy cell

6398 Clin Cancer Res; 17(20) October 15, 2011

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Anti-CD22 Recombinant Immunotoxins

leukemia (HCL), chronic lymphocytic leukemia (CLL), necessary to release domain III from the cell-binding B-cell non-Hodgkin lymphoma (NHL), and acute lym- domain, and domain III contains the ADP-ribosylating phoblastic leukemia (ALL; ref. 15). To kill these cells, we activity that inactivates elongation factor 2. Domain Ib, initially constructed an immunotoxin that reacts with CD22 which has no known function, can be partially or totally by chemically conjugating a toxin fragment of PE to the LL2 removed without affecting toxin activity. In recombinant antibody (16). In studies with this immunotoxin as well as immunotoxins targeting CD22, domain Ia of PE is removed several other chemical conjugates, we found that these and replaced by the Fv portion of an antibody reacting with conjugates were heterogeneous in composition and difficult CD22 as shown in Fig. 1. to produce. Therefore, we turned to recombinant DNA For cancer treatment, the recombinant immunotoxin techniques to make recombinant immunotoxins, which is administered i.v. so that it can reach all of the cells. After are stable, 60- to 65-kDa, homogeneous chimeric proteins injection, the Fv at the amino terminus binds to CD22 in which the Fv of a mAb is fused to truncated PE. The present on the B-cell malignancy being targeted (17), chimeric gene encoding the recombinant immunotoxin is and the carboxyl terminal lysine residue at position 613 introduced into Escherichia coli, where the recombinant (18) is rapidly removed by plasma carboxypeptidase, protein is produced. generating a protein ending in REDL. Very soon after binding, the recombinant immunotoxin-CD22 complex is Cellular intoxication by recombinant immunotoxins internalized through clathrin-coated pits into the endocytic Crystallographic studies have shown that PE is made up compartment (19), where 2 steps occur: (i) reduction of the of 3 major structural domains (Fig. 1). Domain Ia is the cell- disulfide bond in domain II, and (ii) a furin-catalyzed binding domain, domain II contains a furin site that is cleavage in the middle of domain II, resulting in the

Figure 1. Intoxication of cells by moxetumomab pasudotox. Left, illustration of the structure of PE, PE38, and recombinant immunotoxin moxetumomab pasudotox designed to kill CD22-expressing cells. Right, cartoon showing the steps required for the entry and cell killing by moxetumomab pasudotox and similar recombinant immunotoxins containing PE38. After internalization, PE undergoes proteolysis and disulfide-bond reduction to separate the catalytic domain III from the binding domain Ia (20, 60, 61). PE38 undergoes both removal of the carboxyl terminal lysine residue (18) and processing between residues 279 and 280, resulting in a 37-kDa carboxyl terminal toxin fragment ending in the residues REDL. This fragment is believed to be transported intracellularly via the KDEL receptor from the Golgi to the endoplasmic reticulum (ER; 21), where it translocates to the cytosol, resulting in ADP-ribosylation of elongation factor 2 (9), leading to apoptotic cell death (25).

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6399

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. CCR FOCUS

separation of the Fv from domain III (Fig. 1; ref. 20). The that could be safely achieved in cynomolgus monkeys (38). REDL sequence at the carboxyl terminus of the protein then Primary ALL, CLL, and NHL cells were also tested ex vivo and binds to the KDEL recycling receptor (21), and the ADP- found to be sensitive to BL22 (39). CLL with as few as 350 ribosylating domain is transported to the endoplasmic CD22 sites/cell could be killed (39). Thus, the preclinical reticulum, from which it translocates to the cytosol (22, data justified the testing of BL22 in humans. A Good 23). Once in the cytosol, the toxin catalyzes ADP ribosyla- Manufacturing Practices lot of BL22 was produced at the tion of the diphthamide residue (24) in EF-2 (9). This leads Monoclonal Antibody and Recombinant Protein facility of to a rapid decrease in the levels of the antiapoptotic protein the National Cancer Institute, and clinical trials were initi- Mcl-1 and, with the assistance of BAK, initiates the apoptotic ated in 1999. cascade (25–29). Phase I–II activity of BL22 Development of recombinant immunotoxins for CD22 In the phase I trial, we elected to give BL22 every other day malignancies (QOD), for a total of 3 doses per cycle, and to repeat cycles The first recombinant immunotoxin that was designed to every 21 to 28 days. The QOD dosing was based on animal kill CD22-expressing cells contained a single-chain Fv of the experiments that showed that toxicity generally occurs anti-CD22 RFB4 antibody, which had been previously within 48 hours of each dose. Therefore, keeping the dosing isolated by Peter Amlot in England, fused to PE38 (Fig. 1 interval to 2 days in patients may prevent cumulative and Fig. 2). This recombinant immunotoxin had relatively toxicity, particularly during dose escalation. In targeting modest cytotoxic activity, with IC50s of 2 to 30 ng/mL, HCL, where high CD22 density represents a sink for the which we attributed to the instability of the 2 variable drug, this dosing interval had an additional advantage in domains linked by the (G4S)3 peptide (30). To increase allowing dying cells to clear prior to the next dose. We began stability, we introduced cysteine residues into the light and at 3 mg/kg and determined the maximum tolerated dose heavy chains of the Fv so that a disulfide bond would form (MTD) to be 40 mg/kg 3. In total, we treated 46 patients and stably anchor the light and heavy chains instead of the who had experienced failure of standard therapies for their peptide linker that is used in most single-chain Fvs (Fig. 2; disease, including 31 with HCL, 4 with B-NHL, and 11 with refs. 31–36). This protein was named BL22, for B-cell B-CLL (40, 41). In the 31 patients with HCL, 19 CRs (61%) leukemia/lymphoma/CD22 (37). and 6 partial remissions (PR; 19%) were achieved, for an overall response rate (ORR) of 81%. A hemolytic uremic Development of BL22 syndrome composed of thrombocytopenia, hemolytic ane- In preclinical studies, BL22 was cytotoxic to a wide variety mia, and renal insufficiency was observed in 4 HCL patients, of CD22-expressing cell lines, being 1.5- to 6.7-fold more and it completely resolved after 6 to 10 days of plasmaphe- cytotoxic than the single-chain recombinant immunotoxin resis. Dose-limiting capillary leak syndrome (CLS) was less (30, 37). BL22 induced complete remission (CR) in nude common, appearing in only 1 HCL patient. Pharmacoki- þ mice bearing CD22 lymphoma xenografts at plasma levels netic analysis of BL22 showed a strong sink effect of CD22

Figure 2. Recombinant immunotoxins in use or under development. BL22 was reported in 1997 (37) as RFB4(dsFv)-PE38 and was later called CAT-3888. VL and VH are disulfide-bonded together using engineered cysteine residues replacing Arg44 of VH and Gly100 of VL. In 2002, BL22 was mutated to HA22 (later called CAT-8015 and moxetumomab pasudotox) by changing SSY to THW at positions 100, 100a, and 100b of VH (45; horizontal red bars). In 2009, the deletion mutant HA22-LR was reported where PE amino acids 251–394 were replaced by amino acids 274–284 containing the Furin cleavage site (50). Most recently (in 2011), HA22-LR-8M, a mutant of HA22-LR, was reported to contain 8 mutations: D406A, R432G, R467A, R490A, R513A, E548A, K590S, and Q592A (59).

6400 Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Anti-CD22 Recombinant Immunotoxins

in HCL during the first cycle, caused by disease burden and to 50-fold (45). The improvement in binding affinity was high levels of soluble CD22, both of which decreased with due to a slowing of the off rate, which led to increased response (42). Because 65% of CRs occurred after just 1 cytotoxicity and antitumor activity despite similar pharma- cycle of BL22, the phase II trial was planned to limit BL22 to cokinetics and animal toxicity (46). The new, higher-affinity 1 cycle and re-treat only those patients who had not version of BL22 was initially named HA22 (high-activity achieved recovery of cytopenias to the level needed for CR. BL22). Later, after it was licensed to Cambridge Antibody In 36 phase II patients receiving 1 cycle of BL22 at 40 mg/kg Technologies, it was named CAT-8015, and it is now being QOD 3, there were 9 CRs (25%) and 9 PRs (25%), for an developed by MedImmune as moxetumomab pasudotox. ORR of 50%. With selective re-treatment of 56% of the patients, the final ORR increased to 72%, including 47% Phase I testing of moxetumomab pasudotox CRs (43). The median time to relapse of cytopenias had not At the 2010 American Society of Hematology Annual been reached after nearly 7 years of follow-up. This indi- Meeting (Orlando, FL; December 4–7, 2010), interim cated significant activity in relapsed/refractory HCL with a results were reported for clinical trials with moxetumomab safety profile supporting continued development. Howev- pasudotox in HCL and ALL. Clinical trials are listed in er, because the activity of BL22 was much lower in CLL, ALL, Table 1. Several striking differences between moxetumo- and NHL than in HCL (41, 44) and activity in the more- mab pasudotox and BL22 have been noted. One is that in common malignancies was essential for commercial devel- the HCL trial, dose-limiting toxicity was not observed and opment, we had to switch our clinical efforts to an improved dose escalation was terminated at 50 mg/kg, a dose level immunotoxin. above the MTD for BL22, because response rates were high at all dose levels. In fact, the ORR from an interim analysis of Development of moxetumomab pasudotox, an moxetumomab pasudotox, 79% (47), was similar to the affinity-matured version of BL22 72% ORR reported from the phase II study of BL22 (43). An important variable in the activity of immunotoxins is Another striking finding is that 3 responses (all CRs) were their affinity for the target antigen, which determines how observed in 12 patients with pediatric ALL (48), represent- much immunotoxin will bind to a target cell at any given ing the first time responses of this magnitude were observed concentration and how long the cell-bound immunotoxin with this type of agent in this aggressive disease. will remain on the cell surface. The longer it stays attached to the target protein (CD22), the more likely it is to enter and Interim phase I results in HCL kill the target cell. BL22 has a moderate affinity for CD22 At the 2010 American Society of Hematology Annual (Kd 10 nmol/L). This affinity is sufficient to kill enough Meeting (49), investigators presented findings obtained HCL cells in patients to achieve CR. HCL cells have a median from 32 patients with refractory HCL, all of whom had of 40,000 CD22 sites per cell, ranging from 10,000 to received 2 or more prior courses of purine analogue therapy >100,000. However, BL22 was much less effective in chil- and needed treatment based on established criteria for HCL dren with ALL (4,500 sites/cell) or CLL (1,200 sites/cell). To (i.e., neutrophils < 1000/mm3, platelets < 100,000/mm3, improve the affinity and activity of BL22, we carried out hemoglobin < 10 g/dL, or symptomatic splenomegaly). mutagenesis studies and selected an Fv with a higher bind- Patients received dose levels of 5 to 50 mg/kg QOD 3in ing affinity by antibody phage display. Mutation of 3 a standard dose-escalation phase, with cycles repeated every residues in CDR3 of the heavy chain of the BL22 Fv (residues 4 weeks. Patients were re-treated until they reached 2 cycles 100, 100a, and 100b) from SSY to THW increased affinity by past CR; however, treatment was stopped earlier if progres- 15-fold and cytotoxicity toward HCL and CLL cells by up sive disease or immunogenicity was detected. Surprisingly,

Table 1. Ongoing phase I trials with moxetumomab pasudotox

Diseases Prior therapy needed for eligibility Current location

HCL 2 prior therapies, 2 purine analogue courses, unless NIH response to first course was <2 years CLLa 2 prior therapies, 1 with NIH, SCRI, IU, MUSC, CCCN, CSMC DLBCL, MCL 1 prior therapy with rituximab FL 2 prior therapies, 1 with rituximab ALL (pediatric) 2 prior therapies, prior HSCT allowed NIH, SJ, DF

Abbreviations: CCCN, Comprehensive Cancer Center of Nevada; CSMC, Cedars Sinai Medical Center; DF, Dana-Farber Cancer Institute; DLBCL, diffuse large B-cell lymphoma; HSCT, hematopoietic stem cell transplant; IU, Indiana University; MCL, mantle cell lymphoma; MUSC, Medical University of South Carolina; SCRI, Sarah Cannon Research Institute; SJ, Saint Jude Children's Research Hospital. aListed as a phase I–II trial.

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6401

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. CCR FOCUS

Moxetumomab pasudotox

HA22-LR Figure 3. Structure of HA22-LR-8M. Ribbon structures show the locations of the mutations in HA22-LR-8M and the deletion from HA22 used in HA22-LR and HA22-LR-8M. Adapted from Onda et al. (59).

R490 D406 K590

Q592 HA22-LR-8M

R432 E548

R467 R513

no dose-limiting toxicity was observed in the 32 patients, syndrome was asymptomatic laboratory abnormalities despite the fact that half of the patients enrolled (n ¼ 16) in 2 patients, with peak creatinine of 1.53 to 1.66 mg/dL were at the 50 mg/kg 3 dose level, which was higher and platelet nadir of 111,000 to 120,000/mm3. Major than the MTD of BL22 (41). The most common adverse responses were observed at all dose levels in the 32 patients events, seen in 20% to 50% of patients, were expected from reported, with CRs observed at all dose levels from 10 prior experience with immunotoxins. These included mild to 50 mg/kg QOD 3, and the CR rate at all doses was CLS causing weight gain, hypoalbuminemia, peripheral 31% at the time of interim analysis. The median time to edema, fever, elevated transaminases, fatigue, and head- response was 2.8 months (49). Only 1 of the 14 patients ache. The only evidence of possible hemolytic uremic who achieved CR relapsed in less than 1 year, indicating

6402 Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Anti-CD22 Recombinant Immunotoxins

that responses were durable. In summary, moxetumomab activity, particularly on CLL cells, where HA22-LR had a pasudotox showed high and durable antitumor activity in median 16-fold improved cytotoxic activity compared patients with relapsed/refractory HCL, statistically similar with HA22 and more than 10-fold reduced nonspecific to BL22 but without dose-limiting toxicity, justifying further toxicity to mice (50). The latter finding may be due to loss clinical development toward the goal of approval for this of residues that interact with endothelium or macrophages disease. Moxetumomab pasudotox is also being evaluated and produce liver damage in mice and possibly CLS in in CLL and B-cell lymphomas, but data are not yet available other animals, including humans (50–53). It is possible from those trials. that the smaller size of HA22-LR (51.0 kDa versus 63.3 kDa for HA22) may allow improved tumor penetra- Interim phase I results in ALL tion. Finally, because we could safely give much higher Because pediatric ALL is a much more rapidly growing doses to mice, we were able to achieve much better anti- and aggressive disease than HCL, patients were treated with tumor activity (50). moxetumomab pasudotox QOD for 6 doses, and the cycles were repeated every 21 days (48). A rapid dose-escalation A Moxetumomab Pasudotox Variant with No scheme was used for the low dose levels (5, 10, and 20 mg/kg Immunogenicity in Mice QOD 6), and then a standard dose escalation began at 30 mg/kg QOD 6. Fourteen patients 5 to 22 years of age Although the incidence of antibody formation in (median, 11 years) were enrolled in the trial. All of these patients treated with moxetumomab pasudotox is low patients had been heavily pretreated with 2 to 8 (median 4) and did not prevent CRs in HCL and ALL, further prior regimens, and 7 had previously undergone stem decreases in toxin immunogenicity remain an important cell transplantation. Common toxicities included elevated goal. For patients with solid tumors and normal immune bilirubin, transaminases, and creatinine, and CLS in- systems, immunogenicity rates of 80% to 90% were cluding hypoalbuminemia, proteinuria, hypoxia, and pleu- observed after 1 cycle of 3 doses (54–56). To deimmunize ral effusion. Grade 3/4 CLS was observed in 2 of the first the toxin, efforts to identify and remove both T-cell (57) 7 patients (both treated at 30 mg/kg QOD 3) but not in and B-cell (58) epitopes are underway. To date, more 7 subsequent patients once dexamethasone prophylaxis progress has been made with the B-cell epitope approach was instituted. As mentioned above, of 12 patients who (53, 59). On the basis of the hypothesis that human and were evaluable for response, 3 (25%) achieved CR after 1 mouse B-cell epitopes are similar, we used mice for these to 2 cycles, and 5 (42%) had hematologic improvement studies and identified 7 major epitopes in PE38: 3 in with reduction in circulating blasts. As in the HCL trial, domain II, and 4 in domain III. The epitopes in domain II CRs were observed at dose levels as low as 10 mg/kg. Thus, are removed in HA22-LR (50), which has a deletion of moxetumomab pasudotox has achieved major responses, most of domain II, and the epitopes in domain III were including CRs, in patients with ALL. Due to the aggressive destroyed by mutating 8 different bulky amino acids to nature of the disease and the young age of these patients, alanine, serine, or glycine to produce HA22-LR-8M (59). even a transient CR can be lifesaving as a bridge to trans- We measured the reactivity of HA22-LR-8M with serum plantation. Further clinical development of moxetumomab from patients who made antibodies to HA22, and we pasudotox is proceeding in ALL to determine whether foundthatreactivitywithantiserawasgreatlydecreased higher dose levels and more frequent dosing can improve but not abolished. We are now engaged in studies to responses. identify and remove the remaining human-specific epi- topes. Our data suggest that new immunotoxins with A Protease-Resistant Moxetumomab Pasudotox low immunogenicity will not be needed for the success- ful treatment of B-cell malignancies, because the immune One impediment to the cytotoxic activity of immuno- system is very suppressed. However, they will be useful toxins is that many of the immunotoxin molecules that for the treatment of solid tumors, where neutralizing enter target cells do not reach the endoplasmic reti- antibodies develop in the vast majority of patients after culum and cytosol; instead, they are transferred to lyso- 1 cycle of 3 doses. For HCL in particular, we believe somes and other compartments containing lysosomal the clinical results (both efficacy and safety profile) for enzymes, where they are inactivated by proteolysis (Fig. 1). moxetumomab pasudotox justify further clinical devel- To identify potential lysosomal protease cleavage sites, opment, including a pivotal phase III trial currently investigators treated recombinant immunotoxins with lyso- under design, with the goal of achieving U.S. Food and somal enzymes and identified the cleavage sites by amino Drug Administration approval for the use of moxetumo- acid analysis (50). These studies revealed that all of the mab pasudotox for this disease. major protease sites were clustered in domain II and could be removed, leading to a new lysosomal protease-resistant immunotoxin named HA22-LR (Fig. 2 and Fig. 3). In Disclosure of Potential Conflicts of Interest HA22-LR, all of domain II is deleted except for the 11 amino I. Pastan is an inventor on patents on moxetumomab pasudotox acids that make up the essential furin-processing site. held by the NIH. The authors disclose no other potential conflicts of HA22-LR had several useful properties, including increased interest.

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6403

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. CCR FOCUS

Acknowledgments Grant Support

The authors thank the members of our current clinical team, Rita Min- National Cancer Institute Intramural Program; development of cemoyer, Elizabeth Maestri, Natasha Kormanik, Barbara Debrah, and Sonya BL22 and moxetumomab was supported in part by MedImmune, Duke. We also thank our research team and collaborators Inger Margulies, LLC. Hong Zhou, Evgeny Arons, David FitzGerald, Alan Wayne, Richard Beers, John Weldon, Laiman Xiang, Byungkook Lee, and Masanori Onda. Principal investigators for the ALL trial included Drs. Alan Wayne, Deepa Bhojwani, and Lewis Silverman, and principal investigators for the HCL trial included Received April 12, 2011; revised July 28, 2011; accepted July 29, 2011; Drs. Martin Tallman, Steven Coutre, and Tadeusz Robak. published online October 14, 2011.

References 1. Witzig TE. Radioimmunotherapy for B-cell non-Hodgkin lymphoma. 20. Ogata M, Fryling CM, Pastan I, FitzGerald DJ. Cell-mediated cleavage Best Pract Res Clin Haematol 2006;19:655–68. of Pseudomonas exotoxin between Arg279 and Gly280 generates the 2. Steiner M, Neri D. Antibody-radionuclide conjugates for cancer ther- enzymatically active fragment which translocates to the cytosol. J Biol apy: historical considerations and new trends. Clin Cancer Res Chem 1992;267:25396–401. 2011;17:6406–16. 21. Kreitman RJ, Pastan I. Importance of the glutamate residue of KDEL in 3. Teicher BA, Chari RVJ. Antibody conjugate therapeutics: challenges increasing the cytotoxicity of Pseudomonas exotoxin derivatives and and potential. Clin Cancer Res 2011;17:6389–97. for increased binding to the KDEL receptor. Biochem J 1995;307: 4. LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX. Trastu- 29–37. zumab emtansine: a unique antibody-drug conjugate in development 22. Theuer C, Kasturi S, Pastan I. Domain II of Pseudomonas exotoxin A for human epidermal growth factor receptor 2–positive cancer. Clin arrests the transfer of translocating nascent chains into mammalian Cancer Res 2011;17:6437–47. microsomes. Biochemistry 1994;33:5894–900. 5. Katz J, Janik JE, Younes A. Brentuximab vedotin (SGN-35). Clin 23. Theuer CP, Buchner J, FitzGerald D, Pastan I. The N-terminal Cancer Res 2011;17:6428–36. region of the 37-kDa translocated fragment of Pseudomonas exo- 6. Blanc V, Bousseau A, Caron A, Carrez C, Lutz RJ, Lambert JM. toxin A aborts translocation by promoting its own export after SAR3419: an anti-CD19-maytansinoid immunoconjugate for the treat- microsomal membrane insertion. Proc Natl Acad Sci U S A 1993; ment of B-cell malignancies. Clin Cancer Res 2011;17:6448–58. 90:7774–8. 7. Ricart AD. Antibody-drug conjugates of calicheamicin derivative: 24. Webb TR, Cross SH, McKie L, Edgar R, Vizor L, Harrison J, et al. and . Clin Cancer Diphthamide modification of eEF2 requires a J-domain protein and is Res 2011;17:6417–27. essential for normal development. J Cell Sci 2008;121:3140–5. 8. Williams P, Galipeau J. GM-CSF-based fusion cytokines as ligands for 25. Brinkmann U, Brinkmann E, Gallo M, Pastan I. Cloning and charac- immune modulation. J Immunol 2011;186:5527–32. terization of a cellular apoptosis susceptibility gene, the human homo- 9. Carroll SF, Collier RJ. Active site of Pseudomonas aeruginosa exotoxin logue to the yeast chromosome segregation gene CSE1. Proc Natl A. Glutamic acid 553 is photolabeled by NAD and shows functional Acad Sci U S A 1995;92:10427–31. homology with glutamic acid 148 of . J Biol Chem 26. Keppler-Hafkemeyer A, Kreitman RJ, Pastan I. Apoptosis induced by 1987;262:8707–11. immunotoxins used in the treatment of hematologic malignancies. Int J 10. Van Ness BG, Howard JB, Bodley JW. ADP-ribosylation of elongation Cancer 2000;87:86–94. factor 2 by diphtheria toxin. Isolation and properties of the novel 27. Decker T, Oelsner M, Kreitman RJ, Salvatore G, Wang QC, Pastan I, ribosyl-amino acid and its hydrolysis products. J Biol Chem et al. Induction of caspase-dependent programmed cell death in B-cell 1980;255:10717–20. chronic lymphocytic leukemia by anti-CD22 immunotoxins. Blood 11. Endo Y, Mitsui K, Motizuki M, Tsurugi K. The mechanism of action of 2004;103:2718–26. and related toxic lectins on eukaryotic ribosomes. The site and the 28. Bogner C, Dechow T, Ringshausen I, Wagner M, Oelsner M, Lutzny G, characteristics of the modification in 28 S ribosomal RNA caused by et al. Immunotoxin BL22 induces apoptosis in mantle cell lymphoma the toxins. J Biol Chem 1987;262:5908–12. (MCL) cells dependent on Bcl-2 expression. Br J Haematol 2010; 12. Yamaizumi M, Mekada E, Uchida T, Okada Y. One molecule of 148:99–109. diphtheria toxin fragment A introduced into a cell can kill the cell. Cell 29. Du X, Youle RJ, FitzGerald DJ, Pastan I. Pseudomonas exotoxin 1978;15:245–50. A-mediated apoptosis is Bak dependent and preceded by the degra- 13. Zamboni M, Brigotti M, Rambelli F, Montanaro L, Sperti S. High- dation of Mcl-1. Mol Cell Biol 2010;30:3444–52. pressure-liquid-chromatographic and fluorimetric methods for the 30. Mansfield E, Chiron MF, Amlot P, Pastan I, FitzGerald DJ. Recombi- determination of adenine released from ribosomes by ricin and gelonin. nant RFB4 single-chain immunotoxin that is cytotoxic towards CD22- Biochem J 1989;259:639–43. positive cells. Biochem Soc Trans 1997;25:709–14. 14. Kreitman RJ. Getting plant toxins to fuse. Leuk Res 1997;21:997–9. 31. Chaudhary VK, Queen C, Junghans RP, Waldmann TA, FitzGerald DJ, 15. Nitschke L. CD22 and Siglec-G: B-cell inhibitory receptors with distinct Pastan I. A recombinant immunotoxin consisting of two antibody functions. Immunol Rev 2009;230:128–43. variable domains fused to Pseudomonas exotoxin. Nature 1989; 16. Kreitman RJ, Hansen HJ, Jones AL, FitzGerald DJP, Goldenberg DM, 339:394–7. Pastan I. Pseudomonas exotoxin-based immunotoxins containing the 32. Kreitman RJ, Batra JK, Seetharam S, Chaudhary VK, FitzGerald DJ, antibody LL2 or LL2-Fab0 induce regression of subcutaneous human Pastan I. Single-chain immunotoxin fusions between anti-Tac and B-cell lymphoma in mice. Cancer Res 1993;53:819–25. Pseudomonas exotoxin: relative importance of the two toxin disulfide 17. Kounnas MZ, Morris RE, Thompson MR, FitzGerald DJ, Strickland DK, bonds. Bioconjug Chem 1993;4:112–20. Saelinger CB. The a2-macroglobulin receptor/low density lipoprotein 33. Brinkmann U, Pai LH, FitzGerald DJ, Willingham M, Pastan I. B3(Fv)- receptor-related protein binds and internalizes Pseudomonas exotoxin PE38KDEL, a single-chain immunotoxin that causes complete regres- A. J Biol Chem 1992;267:12420–3. sion of a human carcinoma in mice. Proc Natl Acad Sci U S A 18. Hessler JL, Kreitman RJ. An early step in Pseudomonas exotoxin 1991;88:8616–20. action is removal of the terminal lysine residue, which allows binding 34. Brinkmann U, Reiter Y, Jung SH, Lee B, Pastan I. A recombinant to the KDEL receptor. Biochemistry 1997;36:14577–82. immunotoxin containing a disulfide-stabilized Fv fragment. Proc Natl 19. Du X, Beers R, Fitzgerald DJ, Pastan I. Differential cellular internaliza- Acad Sci U S A 1993;90:7538–42. tion of anti-CD19 and -CD22 immunotoxins results in different cyto- 35. Reiter Y, Brinkmann U, Kreitman RJ, Jung S-H, Lee B, Pastan I. toxic activity. Cancer Res 2008;68:6300–5. Stabilization of the Fv fragments in recombinant immunotoxins by

6404 Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Anti-CD22 Recombinant Immunotoxins

disulfide bonds engineered into conserved framework regions. Bio- of the anti-CD22 immunotoxin moxetumomab pasudotox. Proceed- chemistry 1994;33:5451–9. ings of the 52nd ASH Annual Meeting; 2010 Dec 4–7; Orlando, FL. 36. Reiter Y, Kreitman RJ, Brinkmann U, Pastan I. Cytotoxic and antitumor 3246. activity of a recombinant immunotoxin composed of disulfide-stabi- 49. Kreitman RJ, Tallman MS, Coutre SE, Robak T, Wilson WH, Stetler- lized anti-Tac Fv fragment and truncated Pseudomonas exotoxin. Int J Stevenson M, et al. A phase 1 study of moxetumomab pasudotox, an Cancer 1994;58:142–9. anti-CD22 recombinant immunotoxin. In: Relapsed/refractory hairy 37. Mansfield E, Amlot P, Pastan I, FitzGerald DJ. Recombinant RFB4 cell leukemia (HCL). Updated Results. Proceedings of the American immunotoxins exhibit potent cytotoxic activity for CD22-bearing cells Society of Hematology Annual Meeting. Orlando, FL; December 4–7, and tumors. Blood 1997;90:2020–6. 2010. p. 2516. 38. Kreitman RJ, Wang QC, FitzGerald DJP, Pastan I. Complete regression 50. Weldon JE, Xiang L, Chertov O, Margulies I, Kreitman RJ, FitzGerald of human B-cell lymphoma xenografts in mice treated with recombi- DJ, et al. A protease-resistant immunotoxin against CD22 with greatly nant anti-CD22 immunotoxin RFB4(dsFv)-PE38 at doses tolerated by increased activity against CLL and diminished animal toxicity. Blood cynomolgus monkeys. Int J Cancer 1999;81:148–55. 2009;113:3792–800. 39. Kreitman RJ, Margulies I, Stetler-Stevenson M, Wang QC, FitzGerald 51. Baluna R, Rizo J, Gordon BE, Ghetie V, Vitetta ES. Evidence for a DJP, Pastan I. Cytotoxic activity of disulfide-stabilized recombinant structural motif in toxins and interleukin-2 that may be responsible for immunotoxin RFB4(dsFv)-PE38 (BL22) toward fresh malignant cells binding to endothelial cells and initiating vascular leak syndrome. Proc from patients with B-cell leukemias. Clin Cancer Res 2000;6:1476–87. Natl Acad Sci U S A 1999;96:3957–62. 40. Kreitman RJ, Wilson WH, Bergeron K, Raggio M, Stetler-Stevenson M, 52. Smallshaw JE, Ghetie V, Rizo J, Fulmer JR, Trahan LL, Ghetie MA, et al. FitzGerald DJ, et al. Efficacy of the anti-CD22 recombinant immuno- Genetic engineering of an immunotoxin to eliminate pulmonary vas- toxin BL22 in chemotherapy-resistant hairy-cell leukemia. N Engl J cular leak in mice. Nat Biotechnol 2003;21:387–91. Med 2001;345:241–7. 53. Onda M, Beers R, Xiang L, Nagata S, Wang QC, Pastan I. An immu- 41. Kreitman RJ, Squires DR, Stetler-Stevenson M, Noel P, FitzGerald DJ, notoxin with greatly reduced immunogenicity by identification and Wilson WH, et al. Phase I trial of recombinant immunotoxin RFB4 removal of epitopes. Proc Natl Acad Sci U S A 2008; (dsFv)-PE38 (BL22) in patients with B-cell malignancies. J Clin Oncol 105:11311–6. 2005;23:6719–29. 54. Pai LH, Wittes R, Setser A, Willingham MC, Pastan I. Treatment of 42. Matsushita K, Margulies I, Onda M, Nagata S, Stetler-Stevenson M, advanced solid tumors with immunotoxin LMB-1: an antibody linked to Kreitman RJ. Soluble CD22 as a tumor marker for hairy cell leukemia. Pseudomonas exotoxin. Nat Med 1996;2:350–3. Blood 2008;112:2272–7. 55. Hassan R, Bullock S, Premkumar A, Kreitman RJ, Kindler H, Will- 43. Kreitman RJ, Stetler-Stevenson M, Margulies I, Noel P, Fitzgerald DJP, ingham MC, et al. Phase I study of SS1P, a recombinant anti-mesothe- Wilson WH, et al. Phase II trial of recombinant immunotoxin RFB4 lin immunotoxin given as a bolus I.V. infusion to patients with mesothe- (dsFv)-PE38 (BL22) in patients with hairy cell leukemia. J Clin Oncol lin-expressing mesothelioma, ovarian, and pancreatic cancers. Clin 2009;27:2983–90. Cancer Res 2007;13:5144–9. 44. Wayne AS, Kreitman RJ, Findley HW, Lew G, Delbrook C, Steinberg 56. Kreitman RJ, Hassan R, Fitzgerald DJ, Pastan I. Phase I trial of SM, et al. Anti-CD22 immunotoxin RFB4(dsFv)-PE38 (BL22) for CD22- continuous infusion anti-mesothelin recombinant immunotoxin SS1P. positive hematologic malignancies of childhood: preclinical studies Clin Cancer Res 2009;15:5274–9. and phase I . Clin Cancer Res 2010;16:1894–903. 57. Harding FA, Stickler MM, Razo J, DuBridge RB. The immunogenicity of 45. Salvatore G, Beers R, Margulies I, Kreitman RJ, Pastan I. Improved humanized and fully human antibodies: residual immunogenicity cytotoxic activity toward cell lines and fresh leukemia cells of a mutant resides in the CDR regions. MAbs 2010;2:256–65. anti-CD22 immunotoxin obtained by antibody phage display. Clin 58. Nagata S, Pastan I. Removal of B cell epitopes as a practical approach Cancer Res 2002;8:995–1002. for reducing the immunogenicity of foreign protein-based therapeu- 46. Alderson RF, Kreitman RJ, Chen T, Yeung P, Herbst R, Fox JA, et al. tics. Adv Drug Deliv Rev 2009;61:977–85. CAT-8015: a second-generation pseudomonas exotoxin A-based 59. Onda M, Beers R, Xiang L, Lee B, Weldon JE, Kreitman RJ, et al. A immunotherapy targeting CD22-expressing hematologic malignan- recombinant immunotoxin against B cell malignancies with no immu- cies. Clin Cancer Res 2009;15:832–9. nogenicity in mice by removal of B-cell epitopes. Proc Natl Acad Sci 47. Kreitman RJ, Tallman MS, Coutre SE, Robak T, Wilson WH, Stetler- U S A 2011;108:5742–7. Stevenson M, et al. Phase 1 trial of recombinant immunotoxin CAT- 60. Chiron MF, Fryling CM, FitzGerald DJ. Cleavage of Pseudomonas 8015 (HA22) in multiply relapsed hairy cell leukemia. J Clin Oncol exotoxin and diphtheria toxin by a furin-like enzyme prepared from 2010;28(May 20 Suppl):6523. beef liver. J Biol Chem 1994;269:18167–76. 48. WayneA,BhojwaniD,RichardsK,Stetler-Stevenson M, Silverman 61. Fryling C, Ogata M, FitzGerald D. Characterization of a cellular pro- LB, Jeha S, et al. Complete remissions in 3 of 12 patients with tease that cleaves Pseudomonas exotoxin. Infect Immun 1992;60: pediatric acute lymphoblastic leukemia (ALL) during phase I testing 497–502.

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6405

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Antibody Fusion Proteins: Anti-CD22 Recombinant Immunotoxin Moxetumomab Pasudotox

Robert J. Kreitman and Ira Pastan

Clin Cancer Res 2011;17:6398-6405.

Updated version Access the most recent version of this article at: http://clincancerres.aacrjournals.org/content/17/20/6398

Cited articles This article cites 59 articles, 40 of which you can access for free at: http://clincancerres.aacrjournals.org/content/17/20/6398.full#ref-list-1

Citing articles This article has been cited by 27 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/17/20/6398.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/17/20/6398. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research.