Immunotoxin Therapy for Lung Cancer.
L. Xie,M. Fan,Z. Zhang,V. Chandramohan,I. Pastan,D. D. Bigner,X. Bao
DOI: https://doi.org/10.4103/0366-6999.200540
2017-01-01
Abstract:INTRODUCTION Lung cancer is the leading cause for cancer-related deaths in both genders throughout the world. In the United States alone, there were 224,390 estimated new lung cancer cases and 158,080 estimated deaths in 2016.[1] As conventional chemotherapy has reached a plateau of effectiveness in lung cancers and fails in those tumors whose growth and metabolism can hardly be distinguished from normal tissues, innovative therapeutic strategies have been explored. The use of novel agents, for example, immunotherapies, has started to show promising potential in the field. Among immunotherapeutic agents, immunotoxins are a class of antibody-conjugated agents that have been developed for clinical application in many malignancies. Immunotoxins usually contain a bacterial or plant toxin payload to induce cell killing, and a targeting domain with an antibody or its fragment to achieve specific binding capacity. Immunotoxins exert the function of cell killing by inhibiting protein synthesis, which is toxic to both dividing and nondividing cells.[2] In this article, we summarize the properties of the commonly used immunotoxins and their application in recent preclinical and clinical studies in lung cancer. Immunotoxin-based therapeutics Immunotoxin refers to a toxin with the targeting part being either an intact monoclonal antibody (mAb) or its fragment. The main function of an immunotoxin is to target specific cell surface molecules using a cytotoxic agent, which can be internalized to induce cell death by protein synthesis inhibition. In general, the essential parts of an immunotoxin are the binding unit (mAb or its fragment) and the cytotoxic unit (engineered toxin), which can be recombinantly linked together. Since immunotoxins function by directly killing the cells instead of inhibiting receptor-mediated signaling pathways, there can be less chance for tumor cells to upregulate rescue mutations or alternative signaling pathways to resist the immunotoxin therapy.[3] Plant toxins can be obtained from nature in the form of holotoxins and hemitoxins. Holotoxins contain a binding domain and an enzymatic domain linked by a disulfide bond. These toxins include ricin, modeccin, mistletoe lectin, and abrin. Compared to holotoxins, hemitoxins contain an enzymatic domain without a binding domain, which includes pokeweed antiviral protein, gelonin, and saporin.[4] It has been proven that both holotoxins and hemitoxins are able to remove the base of A4324 in 28s rRNA so as to preclude the combination of elongation factor (EF)-1 and - 2 with the 60s ribosomal subunit.[56] Bacterial toxins are somewhat different. An important requirement of fusion toxins is that the catalytic domain has to be separated from the other parts intracellularly. The two commonly engineered bacterial toxins are Pseudomonas exotoxin (PE) and diphtheria toxin (DT).[7] Both consist of three functional domains that can be produced as single polypeptide chains. The binding domain, Domain I, is located at the N-terminus, while domains II and III are located at the C-terminus. Domain II has translocation activity. Domain III catalyzes adenosine diphosphate-ribosylation and EF2 inactivation, to inhibit protein synthesis and ultimately causes cell death.[78] It is reported that up to 300 ribosomes can be irrecoverably inhibited in 35 minutes by a single toxin molecule, which is toxic enough to destroy a cancer cell.[9101112] After binding to the specific receptor, internalization of PE occurs through clathrin-coated pits into the endocytic compartment, and then PE is proteolytically cleaved in between amino acid (AA) 270 and 280, with the reduction of the disulfide bond connecting residues 265 and 287, which results in a fragment of 37,000 (AA280-612) at the C-terminus. With the translocation domain, the fragment is then transported to endoplasmic reticulum (ER), from which the catalytic domain is released into cytosol to ribosylates EF-2, leading to its inactivation. DT's killing mechanism is similar but has fewer steps: from the endocytic compartment, DT directly goes into cytosol to function. DT has a different AA sequence with a different enzymatic domain at the N-terminus.[213] Development of immunotoxins In the early 1980s, as mAbs began showing promise in the field of cancer therapy, Blythman et al.[14] first reported a novel immunotoxin that could kill cancer cells. However, the first-generation immunotoxin chemically conjugated a whole toxin to mAbs, which failed to distinguish the target of cancer cells from normal cells due to multiple potential chemical conjugation sites and the existence of the cell-binding domain of a whole toxin, showing unfavorable results in animal models.[15] The second-generation immunotoxins removed the cell-binding domain from the toxin part, thus affecting a much smaller amount of normal cells in animal models.[7] Nevertheless, the products were expensive for manufacturing, and not efficient enough to penetrate large and heterogeneous tumors, although the potency was proven in this generation of immunotoxins.[2] The third and latest generations of immunotoxins are designed to contain only the variable fragment (Fv) portion of a mAb for binding and the translocation and catalytic domains of toxins to kill tumor cells. The current production method for immunotoxins is to massively and cost-effectively use Escherichia coli. A disulfide bond or a peptide linker is engineered to link the heavy and light chain of the antibodies to form a single-chain variable fragment (scFv) or disulfide-stabilized scFv (scdsFv).[27] To date, more novel immunotoxins are increasingly developed with features such as stronger potency, higher affinity and specificity, and less immunogenicity. Improving potency, affinity, and specificity There are several main methods to improve the potency of immunotoxins, which include changing or mutating the toxin structure, assembling different fragments of antibodies, and changing the conjugation between the two parts.[8] Point mutation techniques are used in remodeling original toxins.[16] The binding domain of the toxin is removed or mutated to be non-effective, which results in much smaller constructs, such as PE38 (AA253-364 and AA381-613) and DT388 or DAB389 (the first 388 AA).[17181920] When the construct PE38 translocates into cytosol, it transforms to components including AA280-364 and AA381-613 with only one cysteine residue at position 287. Moreover, with the modification of the antibody from full size to scFv, the tumor penetra tion is further improved, leading to increased access of the tumor mass.[21] Nevertheless, a reduced binding stability is found in this altered form, which leads to the application of an intrachain disulfide bond connecting only the two Fvs of immunoglobulin (heavy-chain variable domain and light-chain variable domain), to maintain stability and affinity.[3] In addition, PE obtained increased potency with the carboxyl terminus sequence REDLK replaced with KDEL. The KDEL residue improves the cytotoxicity of PE by increasing binding to a sorting receptor that retrogradely transports the toxin from the trans-Golgi apparatus to the ER.[3] Decreasing immunotoxin immunogenicity One of the biggest challenges for immunotoxin therapy is its potential immunogenicity, which can originate from either antibody part or toxin part.[16] To avoid the generation of host anti-murine antibodies against the antibody part of an immunotoxin, this part can be further humanized or replaced by a fully human counterpart.[22] As for the more immunogenic toxin domain, it can be reengineered by combining mutations that decrease lymphocyte epitopes to significantly minimize the immunogenicity induced by a foreign toxin protein.[23] Immunotoxins with mutations at both B- and T-cell epitopes can theoretically eliminate the issue of immunogenicity. Mazor et al.[23] engineered the mesothelin-targeting immunotoxin, LMB-T14, for patients with lung cancer by removing both B- and T-cell epitopes to achieve reduced immunogenicity while maintaining cytotoxicity. In addition, studies using immunosuppressant regimens along with the immunotoxins also showed some benefit.[32425] Other regimens, including a lymphocyte-depleting regimen, which consists of pentostatin and cyclophosphamide, were also found promising in delaying the stimulation of neutralizing anti-immunotoxin antibodies, thus allowing repetitive immunotoxin treatments for patients with solid tumors.[25] Pentostatin and cyclophosphamide selectively suppress the effect of T- and B-cells while largely sparing myeloid cells.[26] Reducing adverse effects Immunotoxin-induced toxicity is either targeted or nonspecific. Vascular leak syndrome (VLS) is a typical nonspecific toxicity, which is caused by endothelial cell damage from a high concentration of immunotoxins. In this case, capillaries are injured with fluid leakage. Fluid retains in tissues and causes edema in tissues, and serum albumin level falls. VLS can usually be managed by adequate hydration.[16] It is reported that high-dose ricin-based immunotoxins induce severe vascular collapse.[2] Bacterial toxins may be better in terms of VLS, given the fact that compared to ricin toxin A chain (RTA) that can directly binds to endothelial cells, a ligand is required for modified PE to connect with the endothelium. In an animal model, a mutation on RTA can decrease the occurrence of VLS.[27] Another inducement of toxicity is the unpredicted target effect due to same-target antigens also expressed on normal tissues. It has been reported that if organs with crucial functions, including the liver, neurons, and kidneys, express the same antigens targeted by immunotoxins, they will undergo immunotoxin-induced injury.[282930] Thus, the selected target antigen should be highly specified to avoid targeting normal cells.[2] APPLICATION IN LUNG CANCER TREATMENT Although there are accumulating data of immunotoxins targeting hematologic tumors, solid tumors (e.g., lung cancer) are much more difficult to treat. The tumor cells are highly condensed with tighter junctions in between cells. Furthermore, some researchers suggest that patients with these cancers are less immunosuppressed, less likely to become immunosuppressed with systemic treatment, and more likely to derive neutralizing antibodies to immunotoxins.[27] Immunotoxin therapy for nonsmall cell lung cancer MAb L6 is an immunotoxin that targets antigens expressed on human lung, breast, colon, and ovarian cancers. The antibody is chemically conjugated to the whole structure of ricin. In mice studies, mAb L6 showed cell-killing effects in xenograft human lung adenocarcinoma.[31] Mesothelin has a strong expression in many solid tumors, including lung adenocarcinoma and mesothelioma, but has low expression in mesothelium.[32333435363738394041] SS1P is an immunotoxin combining the SS1 anti-mesothelin antibody and PE38. It is currently combined with pentostatin and cyclophosphamide in a phase II study for immune depletion to reduce its immunogenicity in patients with lung adenocarcinoma and other mesothelin-positive cancers (NCT01362790).[42] The pilot study of SS1P showed that 3 of 10 treatment-refractory mesothelioma patients had major responses persisting more than 18 months.[25] The immunotoxin RG7787, combined with the humanized Fv fragment of SS1 and a modified PE fragment, has been reported to decrease tumor size in a xenograft mesothelin-expres sing lung model.[1643] High expression of the Lewis Y antigen (Ley) is found in many epithelial tumors. For instance, the Ley antigen was found to be expressed in 80% of lung adenocarcinomas and 42% of squamous cell lung carcinomas in an immunohistochemistry analysis.[44] LMB-1 is an immunotoxin of mAb B3 (which reacts with the Ley antigen) and PE38.[45] It is effective in colon cancer and breast cancer patients, with toxicity due to limited specificity involving endothelial cells.[46] Based on LMB-1, its derivative, LMB-9, yet was developed by combining scdsFv of mAb B3 and PE38, which was utilized to treat several different types of advanced solid tumors including recurrent nonsmall cell lung cancer (NSCLC) expressing the Ley antigen (https://clinicaltrials.gov/ct2/show/NCT00019435). However, results from a phase I study did not show significant effectiveness.[247] Naptumomab estafenatox, also known as ABR-217620, is an immunotoxin consisting of the fragment of the antigen-binding part of a mAb targeting 5T4 and the superantigen Staphylococcal enterotoxin A. Over 95% of tumors from patients with NSCLC, renal cancer, and pancreatic cancer have the expression of the 5T4 antigen. Thirty-one patients, including 19 NSCLC patients, were enrolled in a phase I study and had moderate and tolerable side effects. Most patients in this study achieved stable disease.[48] Results from the updated MONO study as well as another phase I study combining docetaxel (COMBO study) revealed that 36% and 38% of patients in the MONO study (with 51% of those being NSCLC patients) and the COMBO study (with 100% of those being NSCLC patients), respectively, reached stable disease (15% had a partial response in COMBO) at a 2-month follow-up with a maximum-tolerated dose of 26 µg/kg and 22 µg/kg, respectively.[49] Immunotoxin therapy for small cell lung cancer Similarly, immunotoxins based on mAbs SWA11 and SWA20, which target human small cell lung cancer (SCLC) antigen clusters w4 and 5A, respectively, conjugated with RTA, has shown promising activity against SCLC in mice.[50515253] The mouse mAb BrE-3 targeting the polypeptide core of antigen MUC1[54] is combined with RTA to form another immunotoxin, which has been reported to be effective in SCLC.[55] CD56, an antigen of the neural cell adhesion molecule family, is the SCLC cluster 1 antigen. The immunotoxin N901-bR, fused by the anti-CD56 antibody N901 and modified ricin, was reported to be potent against SCLC expressing CD56.[56] In a phase I trial, N901-bR was administered in a group of 21 relapsed or refractory SCLC patients. One partial response was reported.[575859] However, the future use of this drug is limited by the results of a phase II study using the same regimen, in which one fatal progressive VLS was found and all patients developed anti-immunotoxin antibodies, despite one stable disease and one complete remission for 3–4 months.[60] HuD is a neuronal RNA-binding protein detected in all SCLC cells. Ehrlich et al.[61] assembled a type of immunotoxin (BW-2) containing the mouse anti-human-HuD mAb and streptavidin/saporin complexes. It was reported that the intratumoral injection of immunotoxins decreased the local tumor progression in six xenograft mouse models of human SCLC without toxicity.[6162] FUTURE DIRECTIONS Improvements in therapeutic techniques that focus on the specific targeting potency and adverse effects of immunotoxins will be useful for lung cancer treatment. First, it is critical to identify new specific tumor antigens on lung cancer cells that can become potential targets. Fortunately, previous studies have revealed many tumor antigens expressed on lung cancer cells that can be potential targets,[6364] including glycoproteins such as epithelial cell adhesion molecules, carcinoembryonic antigen, mucins, podoplanin (PDPN), and tumor-associated glycoprotein 72; growth and differentiation signaling receptors such as epidermal growth factor receptor (EGFR), human epidermal growth factor receptor (HER) 2, HER3, hepatocyte growth factor receptor, insulin-like growth factor 1 receptor, ephrin receptor A3, and tumor necrosis factor-related apoptosis-inducing ligand receptor 1; and stromal and extracellular matrix antigens such as fibroblast activation protein. Antibodies have been developed in previous studies for these potential targets and can be utilized to synthesize targeting immunotoxins to treat those cancer cells expressing specific antigens. For example, NZ-1 and D2C7 immunotoxins have been developed to specifically target PDPN and EGFR overexpressed on the tumor cell surface, respectively, both of which show a robust antitumor efficacy in the preclinical studies.[6566] Currently, most literature about immunotoxins focuses on hematological malignancies and tumors restricted to a certain area (such as malignant brain tumors) due to adverse effects of systemic administration of immunotoxins (e.g., the immunogenicity of the immunotoxin, off-target toxicity, and VLS).[16] Thus, it is important to optimize the method for safer and more efficient delivery of immunotoxins in lung cancer treatment. As an initial step, orthotopic murine lung cancer models have been established using either human xenograft lung cancer cells or Lewis lung carcinoma cells, building a platform to investigate novel immunotoxins in orthotopic animal models.[6768] Due to the rapid clearance of immunotoxins and potential immunogenicity, immunotoxins are usually administered by locoregional delivery into the tumor site instead of via systemic delivery.[1669] Thanks to the development of therapeutic and imaging techniques, Niu et al.[70] successfully injected antitumor agents percutaneously into the lung tumor site using a fine needle under the guidance of computed tomography without any serious adverse event in patients. With the application of locoregional administration, immunotoxin therapy through intratumoral delivery has already been used to successfully treat patients with glioblastoma to increase the local drug concentration and minimize the systemic toxicity and immunogenicity.[7172] Locoregional administration of immunotoxin therapy for the treatment of lung cancer can now move forward based on modern technique improvements. Although immunotoxin monotherapy has been proven to be effective for the treatment of many malignant tumors, its antitumor efficacy can further be enhanced by the appropriate combination strategies with other agents. Studies have shown that a type of newly developed immunotoxin may have better potency by exerting its cytotoxic moiety effects based on human-derived endogenous proteins, such as pro-apoptotic proteins or RNase.[73] Sensitivity of cancer cells to apoptosis will largely affect the cytotoxicity of these sorts of immunotoxins, and inactivation of p53 and upregulations of apoptosis inhibitors (e.g., B-cell lymphoma [Bcl]-2 and Bcl-xL) will lead to drug resistance. Thus, these immunotoxins may be more effective if combined with small molecule inhibitors of anti-apoptotic proteins to sensitize cancer cell apoptosis.[73] Besides, Leshem et al.[74] reported that the combination therapy of RG7787 immunotoxin with anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA4) antibody (an immune checkpoint inhibitor) led to a high rate of complete remissions in their breast cancer model, indicating that the combinatorial therapy of immunotoxin and immune checkpoint inhibitors may promote the activation of the antitumor immunity to achieve a long-term tumor elimination. Currently, two anti-programmed cell death protein 1 (PD-1) immune checkpoint inhibitors, nivolumab and pembrolizumab, were approved by the US Food and Drug Administration (FDA) to treat NSCLC, which can be potential candidates to be combined with immunotoxins to treat advanced NSCLC in the future. In conclusion, the latest immunotoxins have emerged from many studies involving engineered immunotoxins that bind to tumor-surface epitopes with reduced in vivo toxicity and immunogenicity. In many preclinical and clinical studies, immunotoxins have displayed a different mechanism of tumor cell killing than traditional chemotherapy or radiation therapy. Further progress and improved clinical response of immunotoxin therapy against lung cancer depends on the identification of new tumor targets and optimized administration methods to promote its specificity and potency while minimize the adverse effect. Furthermore, immunotoxins may synergistically work with other therapeutics to enhance the antitumor efficacy as a combinatorial therapy. Financial support and sponsorship The work was supported by grants from the Young Scientists Fund from the National Natural Science Foundation of China (No. 81401896), and the Pujiang Talent Program from Shanghai Municipal Human Resource Bureau and Shanghai Science and Technology Committee (No. 14PJ1402000). Conflicts of interest Darell D Bigner owns stock in Istari Oncology and is a consultant to Genetron Health. Acknowledgment We would like to thank Jenna Lewis for editing our manuscript.
What problem does this paper attempt to address?
-
Immunotoxins: Targeted Toxin Delivery for Cancer Therapy
Xin Mei,Junsheng Chen,Jing Wang,Jianwei Zhu
DOI: https://doi.org/10.1055/s-0039-1700507
2019-01-01
Pharmaceutical Fronts
Abstract:Immunotoxins are proteins that consist of a protein toxin conjugated to a specific targeting moiety. The targeting moiety is usually an antibody or ligand, such as monoclonal antibody, antibody fragment, a cytokine, or a growth factor. The toxins usually come from plant toxins, bacterial toxins, or human-origin cytotoxic elements. Nearly all toxins work by enzymatically inhibiting protein synthesis. After binding to antigens or receptors on target cell surfaces, immunotoxins are internalized and translocated to the cytosol where they can kill the cells. Immunotoxins have demonstrated high cytotoxicity to cancer cells and to date two immunotoxins have been approved by U.S. Food and Drug Administration on the markets for the treatment of hematological tumors: Lumoxiti and Ontak. Many other molecules are under development or clinical trials for different forms of cancer. Although immunotoxins exhibit great potency in xenograft model systems and early clinical trials, there are obstacles that limit successful treatments, including immunogenicity, nonspecific toxicity, and poor penetration. However, efforts are underway to address these problems. In this review, we summarize immunotoxins currently in clinical trials for either hematological tumors or solid tumors, outline the design of immunotoxins utilizing variety of components, and discuss the prominent examples of redesigned immunotoxins with reduced immunogenicity and nonspecific toxicity, as well as the strategies in manufacturing immunotoxins. With further improvements, it is anticipated that immunotoxins will play an increasing role in cancer therapy.
-
HER2-targeted immunotoxins with low nonspecific toxicity and immunogenicity
Rui Guo,Li Cao,Wenjun Guo,Hui Liu,Hua Xu,Qi Fang,Zhangyong Hong
DOI: https://doi.org/10.1016/j.bbrc.2016.05.044
2016-06-17
Abstract:Immunotoxins have efficient anti-tumor activity due to their extreme potency. However, dose-limiting off-target toxicity and immunogenicity are the critical barriers for these immunotoxins to be used in a clinical setting. In this study, we designed a Pseudomonas exotoxin A (PE)-based human epidermal growth factor receptor-2 (HER2)-specific immunotoxin HER2-PE25-X7 by deleting most of domain II and introducing seven point mutations into domain III of the PE38 toxin. The anti-cancer activity, off-target toxicity and immunogenicity of this immunotoxin were carefully evaluated in vitro and in vivo. This new construct maintained the therapeutic potency of the original PE38-based immunotoxin HER2-PE38, with a greatly reduced off-target toxicity and immunogenicity. To compare with HER2-PE38, which resulted in the death of most of the mice after a single dose of 1.0 mg/kg, the new construct was completely tolerated at a dose of 10 mg/kg by the mice and almost completely depleted the tumor after treatment with five doses of 5 mg/kg of the immunotoxin. This work demonstrates a potentially attractive therapeutic modality for HER2-specific cancer treatment.
-
Immunotoxins and Cancer Therapy
Zheng Li,Tao Yu,Ping Zhao,Jie Ma
2005-01-01
Abstract:In the past decade, an increased amount of clinically-oriented research involving immunotoxins has been published.Immunotoxins are a group of artificially-made cytotoxic molecules targeting cancer cells. These molecules composed of a targeting moiety, such as a ligand or an antibody, linked to toxin moiety, which is a toxin with either truncated or deleted cell-binding domain that prevents it from binding to normal cells. Immunotoxins can be divided into two categories: chemically conjugated immunotoxins and recombinant ones. The immunotoxins of the first category have shown limited efficacy in clinical trials in patients with hematologic malignancies and solid tumors. Within the last few years, single-chain immunotoxins provide enhanced therapeutic efficacy over conjugated forms and result in improved antitumor activity. In this review, we briefly illustrate the design of the immunotoxins and their applications in clinical trials. Cellular & Molecular Immunology. 2005;2(2):106-112.
-
Pseudomonas Exotoxin-Based Immunotoxins: Over Three Decades of Efforts on Targeting Cancer Cells With the Toxin
Seyed Mehdi Havaei,Marc G. Aucoin,Ali Jahanian-Najafabadi
DOI: https://doi.org/10.3389/fonc.2021.781800
IF: 4.7
2021-12-16
Frontiers in Oncology
Abstract:Cancer is one of the prominent causes of death worldwide. Despite the existence of various modalities for cancer treatment, many types of cancer remain uncured or develop resistance to therapeutic strategies. Furthermore, almost all chemotherapeutics cause a range of side effects because they affect normal cells in addition to malignant cells. Therefore, the development of novel therapeutic agents that are targeted specifically toward cancer cells is indispensable. Immunotoxins (ITs) are a class of tumor cell-targeted fusion proteins consisting of both a targeting moiety and a toxic moiety. The targeting moiety is usually an antibody/antibody fragment or a ligand of the immune system that can bind an antigen or receptor that is only expressed or overexpressed by cancer cells but not normal cells. The toxic moiety is usually a protein toxin (or derivative) of animal, plant, insect, or bacterial origin. To date, three ITs have gained Food and Drug Administration (FDA) approval for human use, including denileukin diftitox (FDA approval: 1999), tagraxofusp (FDA approval: 2018), and moxetumomab pasudotox (FDA approval: 2018). All of these ITs take advantage of bacterial protein toxins. The toxic moiety of the first two ITs is a truncated form of diphtheria toxin, and the third is a derivative of Pseudomonas exotoxin (PE). There is a growing list of ITs using PE, or its derivatives, being evaluated preclinically or clinically. Here, we will review these ITs to highlight the advances in PE-based anticancer strategies, as well as review the targeting moieties that are used to reduce the non-specific destruction of non-cancerous cells. Although we tried to be as comprehensive as possible, we have limited our review to those ITs that have proceeded to clinical trials and are still under active clinical evaluation.
oncology
-
Study on the Preparation of the Immunotoxin 2E8-NCTD and Its Targeting Killing Effect in Vitro.
Yongmin Tang,Lixia Li,Di Wang,Hongqiang Shen,Baiqin Qian,Chunfang Luo,Haizhong Zhang
DOI: https://doi.org/10.1182/blood.v110.11.4186.4186
IF: 20.3
2007-01-01
Blood
Abstract:OBJECTIVE:Monoclonal antibody (mAb) conjugated with certain toxin to generate immunotoxin bears an important and promising effect as a new therapy for patients with hematopoietic malignancies. However, most toxic moieties conjugated with antibody proteins reported in the literature were toxic proteins which presented immunogenicity to patients capable of inducing anti-toxin antibody. Norcantharidin (NCTD) is a small molecule toxin. It does not have the immunogenicity to human body so that it bears a promising potential for development of new targeting drug. In this study, a new clone of self-made anti-CD19 mAb named ZCH-4-2E8 conjugated with NCTD was used to investigate its targeting efficacy against CD19+ lymphoid malignant Nalm-6 cells in vitro to provide the experimental data for the further development of this new targeting agent.METHODS:A monoclonal antibody named 2E8 was prepared from mouse ascites and purified by gel chromatography. The purity of the antibody protein was checked by SDS-PAGE assay. Immunotoxin 2E8-NCTD was successfully generated through conjugating CD19 mAb protein and Norcantharidin by the activated ester method. The binding activity of the immunoconjugate (2E8-NCTD) against CD19 antigens on cell surface and the expression levels of CD19 antigens on Nalm-6 and K562 cells were examined by flow cytometry. Comparisons of the inhibitory effects among PBS, purified 2E8 antibody, norcantharidin and immunotoxin 2E8-NCTD groups on cell growth of either Nalm-6 cells or K562 cells were made.RESULTS:The purity of the purified 2E8 antibody was higher than 99.00% demonstrated by SDS-PAGE assay. 2E8 antibody in the supernatant reacted with 99.34% of Nalm-6 cells, while only 0.98% of K562 cells reacted with this antibody. The newly generated immunotoxin (2E8-NCTD) had a positive rate of 99.90% on Nalm-6 cells with little reduction of binding activity. From the in vitro study, both 2E8-NCTD and norcantharidin were shown to have significant inhibitory effects on the growth of CD19+ Nalm-6 cells (P < 0.001), while the purified 2E8 antibody did not show any significant influences on the growth of Nalm-6 cells. Since no significant inhibitory effects were identified among immunotoxin 2E8-NCTD, 2E8 antibody and control groups on CD19(-) K562 cells, a significant targeting effect of the 2E8-NCTD against Nalm-6 cells was confirmed.CONCLUSIONS:The immunotoxin 2E8-NCTD was successfully synthesized by activated ester method with an excellent targeting killing effect on CD19+ Nalm-6 leukemia cells in vitro, which provides some experimental data for the further development of this new targeting agent.
-
Preparation of Diphtheria and Pseudomonas Exotoxin A Immunotoxins and Evaluation of Their Cytotoxicity Effect on SK-BR-3, BT-474, and MDA-MB-231 Breast Cancer Cell Lines
Sahel Amoozadeh,Maryam Hemmati,Mohammad Morad Farajollahi,Neda Akbari,Parastoo Tarighi
DOI: https://doi.org/10.1080/07357907.2019.1655761
Abstract:Immunotoxin targeted therapy is a promising way of cancer therapy that is made from a toxin attached to an antibody which target a specific protein presented on cancer cells. In this study, we introduce immunotoxins comprising of truncated pseudomonas exotoxin A (PEA) and diphtheria toxin (DT) conjugated to trastuzumab. The effectiveness of 20 and 30 μg/ml immunotoxins and trastuzumab were studied on SK-BR-3 and BT-474 HER2/neu positive breast cancer cell lines by a cell death assay test. The produced immunotoxins have the potential to reduce the therapeutic dose of the trastuzumab and in the same time achieve higher efficiency.
-
Generation of Potent Anti-HER1/2 Immunotoxins by Protein Ligation Using Split Inteins
Thomas Pirzer,Kira-Sophie Becher,Marcel Rieker,Tobias Meckel,Henning D Mootz,Harald Kolmar
DOI: https://doi.org/10.1021/acschembio.8b00222
2018-08-17
Abstract:Cell targeting protein toxins have gained increasing interest for cancer therapy aimed at increasing the therapeutic window and reducing systemic toxicity. Because recombinant expression of immunotoxins consisting of a receptor-binding and a cell-killing moiety is hampered by their high toxicity in a eukaryotic production host, most applications rely on recombinant production of fusion proteins consisting of an antibody fragment and a protein toxin in bacterial hosts such as Escherichia coli ( E. coli). These fusions often lack beneficial properties of whole antibodies like extended serum half-life or efficient endocytic uptake via receptor clustering. Here, we describe the production of full-length antibody immunotoxins using self-splicing split inteins. To this end, the short (11 amino acids) N-terminal intein part of the artificially designed split intein M86, a derivative of the Ssp DnaB intein, was recombinantly fused to the heavy chain of trastuzumab, a human epidermal growth factor receptor 2 (HER2) receptor targeting antibody and to a nanobody-Fc fusion targeting the HER1 receptor, respectively. Both antibodies were produced in Expi293F cells. The longer C-terminal counterpart of the intein was genetically fused to the protein toxins gelonin or Pseudomonas Exotoxin A, respectively, and expressed in E. coli via fusion to maltose binding protein. Using optimized in vitro splicing conditions, we were able to generate a set of specific and potent immunotoxins with IC50 values in the mid- to subpicomolar range.
-
Advances in immunotoxin engineering: precision therapeutic strategies in modern oncology
Akbar Oghalaie,Mahmoud Eshagh Hosseini,Mohammad Hosseininejad-Chafi,Zohre Eftekhari,Mahdi Behdani,Fatemeh Kazemi-Lomedasht
DOI: https://doi.org/10.1007/s12032-024-02478-3
2024-09-04
Abstract:Immunotoxins (ITs) are specialized therapeutic agents designed for targeted treatment, particularly in cancer therapy. They consist of a monoclonal antibody or antibody fragment linked to a potent cytotoxic agent, such as bacterial- or plant-derived toxins like diphtheria toxin, ricin, or pseudomonas exotoxin. The monoclonal antibody component specifically binds to antigens expressed on the surface of target cells, facilitating the internalization of the IT. Once inside the cell, the cytotoxic agent is released, disrupting essential cellular processes and leading to cell death. This targeted approach minimizes damage to healthy tissues while effectively eliminating diseased cells. The production of ITs involves two primary methods: recombinant fusion and chemical conjugation. In recombinant fusion, genetic engineering is used to create a fusion protein that combines the antibody and toxin, ensuring precise control over their ratio and functionality. In chemical conjugation, pre-existing antibodies are chemically linked to toxins, allowing for greater flexibility in combining different antibodies and cytotoxic agents. Each method has its advantages and challenges, influencing the specificity, production complexity, and therapeutic potential of the resulting ITs. As research advances, ITs continue to show promise not only in oncology but also in treating other diseases, including inflammatory conditions and atherosclerosis. The precise targeting and potent effects of ITs make them a valuable tool in the development of new therapeutic strategies.
-
Target-specific cytotoxic effects on HER2-expressing cells by the tripartite fusion toxin ZHER2:2891-ABD-PE38X8, including a targeting affibody molecule and a half-life extension domain
Hao Liu,Johan Seijsing,Fredrik Y Frejd,Vladimir Tolmachev,Torbjörn Gräslund
DOI: https://doi.org/10.3892/ijo.2015.3027
Abstract:Development of cancer treatment regimens including immunotoxins is partly hampered by their immunogenicity. Recently, deimmunized versions of toxins have been described, potentially being better suited for translation to the clinic. In this study, a recombinant tripartite fusion toxin consisting of a deimmunized version of exotoxin A from Pseudomonas aeruginosa (PE38) genetically fused to an affibody molecule specifically interacting with the human epidermal growth factor receptor 2 (HER2), and also an albumin binding domain (ABD) for half-life extension, has been produced and characterized in terms of functionality of the three moieties. Biosensor based assays showed that the fusion toxin was able to interact with human and mouse serum albumin, but not with bovine serum albumin and that it interacted with HER2 (KD=5 nM). Interestingly, a complex of the fusion toxin and human serum albumin also interacted with HER2 but with a somewhat weaker affinity (KD=12 nM). The IC50-values of the fusion toxin ranged from 6 to 300 pM on SKOV-3, SKBR-3 and A549 cells and was lower for cells with higher surface densities of HER2. The fusion toxin was found specific for HER2 as shown by blocking available HER2 receptors with free affibody molecule before subjecting the cells to the toxin. Analysis of contact time showed that 10 min was sufficient to kill 50% of the cells. In conclusion, all three regions of the fusion toxin were found to be functional.
-
Abstract 1919: Activity and affinity tuning next-generation immunotoxins for targeted therapy
Huazhu (Peter) Liang,Greg L. Beilhartz,Shi Bo Cao,Roman A. Melnyk
DOI: https://doi.org/10.1158/1538-7445.am2024-1919
IF: 11.2
2024-03-22
Cancer Research
Abstract:Abstract Classic cancer-associated receptors, such as EGFR and Her2, are often overexpressed on malignant cells relative to healthy cells. Numerous molecules have been developed to leverage this differential expression with the goal of developing specific and safe targeted therapies, including antibody-drug-conjugates and immunotoxins. The general schematic of these molecules is to fuse toxic payloads -either small molecules or proteins - with a targeting moiety that binds to the cancer-specific receptor. Despite the general success of this approach, even low levels of these receptors on healthy tissues result in on-target/off-tumor toxicity thus limiting the dose of therapeutic molecules available to tumors. Targeted therapies with greater on-tumor efficacy and reduced toxicity are urgently needed in order to further advance therapeutic outcomes. To achieve this, we set out to exploit the unique features of bacterial toxins as highly modular protein delivery systems to design next-generation targeted therapies. We demonstrate the capacity of the immunotoxin platform to deliver various therapeutic proteins, with distinct activities from inhibiting protein synthesis to degrading oncogenic signaling pathways, such as the RAS/MAPK pathway. Further, we show how modifying the affinity, valency, and specificity of the receptor-binding moieties, as well as tuning the enzymatic activity of the toxic enzyme payloads interact to affect the on-target efficacy and off-target toxicity of the targeted molecules. Altogether, these data demonstrate the potential of immunotoxins as highly flexible and tunable platforms for the development of a new class of anti-cancer therapeutics. Citation Format: Huazhu (Peter) Liang, Greg L. Beilhartz, Shi Bo Cao, Roman A. Melnyk. Activity and affinity tuning next-generation immunotoxins for targeted therapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 1919.
oncology
-
Abstract 2661: Engineered Toxin Bodies (ETBs): Clinical stage immunotoxins with a safer and differentiated profile
Chris Moore,Lee Robinson,Garrett Cornelison,Joseph Dekker,Roger Waltzman,John Majercak,Joseph Phillips,Jay Zhao,Jason Kim,Eric Poma
DOI: https://doi.org/10.1158/1538-7445.am2023-2661
IF: 11.2
2023-04-04
Cancer Research
Abstract:Abstract Immunotoxins (the fusion of a bacterial cytotoxic protein with a targeting moiety) have been explored since the 1970s but have been limited in their therapeutic utility because of the propensity to drive a host innate immune response resulting in potentially fatal cytokine elevations and capillary leak syndrome (CLS). Engineered Toxin Bodies (ETBs) represent an important evolution in immunotoxins, overcoming the issues with innate immunity and adding biologic utility not previously seen. ETBs use an antibody-based targeting domain genetically fused to a de-immunized form of the Shiga-like toxin A subunit (SLTA). Like previous immunotoxins, ETBs bind and induce cell-death in a target-specific manner but have additional biology not inherent to other immunotoxins. ETBs can induce their own internalization, allowing for the targeting of poorly/non internalizing receptors. ETBs can deliver other payloads like viral class I antigen or small molecules in addition to SLTA, allowing for multiple MOAs. And, most importantly, we have de-immunized SLTA through genetic engineering to create ETBs devoid of innate immune effects/CLS seen with previous immunotoxins. Three ETBs (MT-0169, MT-5111, and MT-6402) are currently in clinical studies across different targets (CD38, HER2, PD-L1) and across hematologic malignancies, solid tumor, and immuno-oncology indications. To date, no clinical manifestations of CLS have been observed in 80+ patients; in contrast, the rate of CLS for approved immunotoxins ranges for 33% to >50%. In general, all three ETBs have shown good tolerability with little off-target toxicity and all three ETBs have shown monotherapy clinical benefit in heavily pre-treated patients. MT-6402, an ETB targeting PD-L1, is the first ETB to carry the additional mechanism of antigen seeding. Along with its ability to destroy ribosome in a PD-L1-targeted fashion, MT-6402 can deliver a class I antigen derived from human cytomegalovirus (CMV) to the ER, allowing for presentation of the antigen in complex with MHC class I on the cell surface of a tumor cell. Early clinical data demonstrate antigen seeding with MT-6402 can result in T-cells specific to the seeded CMV antigen mounting an immune response to the tumor. ETBs represent a unique and wholly distinct scaffold for drug development in oncology. ETBs allow for the targeting of non-internalizing receptors that are not amenable to ADCs. ETBs have a mechanism of cell-kill (enzymatic and irreversible ribosomal destruction) that is distinct from any approved agent in oncology. ETBs can also deliver additional payloads to drive unique biology like the alteration of tumor immunophenotype. Here we describe three active clinical stage programs with encouraging safety and efficacy data that represent a transformation of the immunotoxin landscape into a more viable therapeutic approach to target validated as well as typically intractable clinical cancer targets. Citation Format: Chris Moore, Lee Robinson, Garrett Cornelison, Joseph Dekker, Roger Waltzman, John Majercak, Joseph Phillips, Jay Zhao, Jason Kim, Eric Poma. Engineered Toxin Bodies (ETBs): Clinical stage immunotoxins with a safer and differentiated profile [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2661.
oncology
-
IgG-Engineered Protective Antigen for Cytosolic Delivery of Proteins into Cancer Cells
Zeyu Lu,Nicholas L. Truex,Mariane B. Melo,Yiran Cheng,Na Li,Darrell J. Irvine,Bradley L. Pentelute
DOI: https://doi.org/10.1021/acscentsci.0c01670
IF: 18.2
2021-02-04
ACS Central Science
Abstract:Therapeutic immunotoxins composed of antibodies and bacterial toxins provide potent activity against malignant cells, but joining them with a defined covalent bond while maintaining the desired function is challenging. Here, we develop novel immunotoxins by dovetailing full-length immunoglobulin G (IgG) antibodies and nontoxic anthrax proteins, in which the C terminus of the IgG heavy chain is connected to the side chain of anthrax toxin protective antigen. This strategy enabled efficient conjugation of protective antigen variants to trastuzumab (Tmab) and cetuximab (Cmab) antibodies. The conjugates effectively perform intracellular delivery of edema factor and N terminus of lethal factor (LF<sub>N</sub>) fused with diphtheria toxin and Ras/Rap1-specific endopeptidase. Each conjugate shows high specificity for cells expressing human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR), respectively, and potent activity across six Tmab- and Cmab-resistant cell lines. The conjugates also exhibit increased pharmacokinetics and pronounced in vivo safety, which shows promise for further therapeutic development.
chemistry, multidisciplinary
-
Improving the cytotoxicity of immunotoxins by reducing the affinity of the antibody in acidic pH
Xiaoyu Liu,Qingqing Tan,Jiaqi Wen,Xufei Wang,Gang Yang,Yuxiao Li,Ming Lu,Wei Ye,Anfeng Si,Sujuan Ma,Tong Ding,Luan Sun,Fang Liu,Mei Zhang,Tao Jiang,Wei Gao
DOI: https://doi.org/10.1186/s12967-023-04210-7
IF: 8.44
2023-08-28
Journal of Translational Medicine
Abstract:Immunotoxins are antibody-toxin conjugates that bind to surface antigens and exert effective cytotoxic activity after internalization into tumor cells. Immunotoxins exhibit effective cytotoxicity and have been approved by the FDA to treat multiple hematological malignancies, such as hairy cell leukemia and cutaneous T-cell lymphoma. However, most of the internalized immunotoxin is degraded in lysosomes, and only approximately 5% of free toxin escapes into the cytosol to exert cytotoxicity. Many studies have improved immunotoxins by engineering the toxin fragment to reduce immunogenicity or increase stability, but how the antibody fragment contributes to the activity of immunotoxins has not been well demonstrated.
medicine, research & experimental
-
Reduction of non-specific toxicity of immunotoxin by intein mediated reconstitution on target cells
Jing Wang,Lei Han,Junsheng Chen,Yueqing Xie,Hua Jiang,Jianwei Zhu
DOI: https://doi.org/10.1016/j.intimp.2018.11.039
Abstract:Recombinant immunotoxins are chimeric proteins composed of a targeting peptide that binds to a specific tumor antigen and a toxin protein killing target cells. Recombinant immunotoxin exhibits potent cancer inhibiting effects both in vivo and in vitro. However, the non-specific toxicity causes severe syndromes limiting their clinical application. To reduce toxicity caused by recombinant immunotoxins in general, we divided an immunotoxin into two nontoxic segments that may restore toxic bioactivity on tumor cell surface based on the intein mediated trans-splicing reaction. Both split and reconstituted immunotoxins were tested for their biological activities. We found that the reconstituted immunotoxin retained antigen specificity and affinity toward cancer cells overexpressing HER2/neu. After being internalized into HER2/neu positive cells, the reconstituted immunotoxin showed comparable cytotoxicity as the original immunotoxin, while the split immunotoxin fragments showed no toxic activity to cells with or without HER2/neu expression. This approach can potentially be used under clinical settings to reduce non-specific toxicity by administering patients with inactive immunotoxin fragments. Cytotoxic effect only occurs at tumor sites where the inactive fragments bind, trans-splice and become active toxin.
-
Modular Conjugation of a Potent Anti-HER2 Immunotoxin Using Coassociating Peptides
Audrey Stoessel,Nadja Groysbeck,Lucile Guyot,Lina Barret,Yves Nominé,Leonel Nguekeu-Zebaze,Ambre Bender,Laetitia Voilquin,Thomas Lutz,Nikita Pallaoro,Marie Blocat,Celia Deville,Murielle Masson,Guy Zuber,Bruno Chatton,Mariel Donzeau
DOI: https://doi.org/10.1021/acs.bioconjchem.0c00482
2020-10-21
Abstract:Immunotoxins are emerging candidates for cancer therapeutics. These biomolecules consist of a cell-targeting protein combined to a polypeptide toxin. Associations of both entities can be achieved either chemically by covalent bonds or genetically creating fusion proteins. However, chemical agents can affect the activity and/or stability of the conjugate proteins, and additional purification steps are often required to isolate the final conjugate from unwanted byproducts. As for fusion proteins, they often suffer from low solubility and yield. In this report, we describe a straightforward conjugation process to generate an immunotoxin using coassociating peptides (named K3 and E3), originating from the tetramerization domain of p53. To that end, a nanobody targeting the human epidermal growth factor receptor 2 (nano-HER2) and a protein toxin fragment from Pseudomonas aeruginosa exotoxin A (TOX) were genetically fused to the E3 and K3 peptides. Entities were produced separately in Escherichia coli in soluble forms and at high yields. The nano-HER2 fused to the E3 or K3 helixes (nano-HER2-E3 and nano-HER2-K3) and the coassembled immunotoxins (nano-HER2-K3E3-TOX and nano-HER2-E3K3-TOX) presented binding specificity on HER2-overexpressing cells with relative binding constants in the low nanomolar to picomolar range. Both toxin modules (E3-TOX and K3-TOX) and the combined immunotoxins exhibited similar cytotoxicity levels compared to the toxin alone (TOX). Finally, nano-HER2-K3E3-TOX and nano-HER2-E3K3-TOX evaluated on various breast cancer cells were highly potent and specific to killing HER2-overexpressing breast cancer cells with IC50 values in the picomolar range. Altogether, we demonstrate that this noncovalent conjugation method using two coassembling peptides can be easily implemented for the modular engineering of immunotoxins targeting different types of cancers.
-
Protection of the Furin Cleavage Site in Low-Toxicity Immunotoxins Based on Pseudomonas Exotoxin A
Gilad Kaplan,Fred Lee,Masanori Onda,Emily Kolyvas,Gaurav Bhardwaj,David Baker,Ira Pastan
DOI: https://doi.org/10.3390/toxins8080217
IF: 5.075
2016-07-25
Toxins
Abstract:Recombinant immunotoxins (RITs) are fusions of an Fv-based targeting moiety and a toxin. Pseudomonas exotoxin A (PE) has been used to make several immunotoxins that have been evaluated in clinical trials. Immunogenicity of the bacterial toxin and off-target toxicity have limited the efficacy of these immunotoxins. To address these issues, we have previously made RITs in which the Fv is connected to domain III (PE24) by a furin cleavage site (FCS), thereby removing unneeded sequences of domain II. However, the PE24 containing RITs do not contain the naturally occurring disulfide bond around the furin cleavage sequence, because it was removed when domain II was deleted. This could potentially allow PE24 containing immunotoxins to be cleaved and inactivated before internalization by cell surface furin or other proteases in the blood stream or tumor microenvironment. Here, we describe five new RITs in which a disulfide bond is engineered to protect the FCS. The most active of these, SS1-Fab-DS3-PE24, shows a longer serum half-life than an RIT without the disulfide bond and has the same anti-tumor activity, despite being less cytotoxic in vitro. These results have significance for the production of de-immunized, low toxicity, PE24-based immunotoxins with a longer serum half-life.
toxicology,food science & technology
-
Immunotoxin-mediated targeting of claudin-4 inhibits the proliferation of cancer cells
S M Hashimi,S Yu,N Alqurashi,D S Ipe,M Q Wei
DOI: https://doi.org/10.3892/ijo.2013.1881
Abstract:Immunotoxins are engineered chimeric proteins that consist of a fragment of a toxin fused to a modified antibody or growth factor capable of targeting specific cells. Furthermore, these proteins can be targeted to receptors that are commonly overexpressed on cancer cells. The majority of immunotoxins function by binding to cells, translocating into the cytosol and inhibiting protein synthesis. In this study, the expression of claudin‑4 (CLDN4) in various cancer cells was analysed as a potential target for immunotoxins. To target CLDN4-expressing cancer cells, the c-terminal CLDN4‑binding domain of Clostridium perfringens enterotoxin (CPE) was fused to the Pseudomonas aeruginosa exotoxin A (ETA) domain to create an immunotoxin (CPE‑ETA'). Subsequently, the capacity of such an immunotoxin in suppressing the proliferation of CLDN4-positive cancer cells was investigated. We report that head and neck squamous carcinoma cells (HN5) have an elevated CLDN4 expression compared to the other cell lines tested. Our findings further demonstrate that CPE‑ETA' is highly potent against MCF-7 breast [50% inhibitory concentration (IC50) 9.8 ng/ml] and HN5 head/neck (IC50 8.8 ng/ml) cancer cell lines, while it has no cytotoxic effects on HeLa cells (CLDN4‑negative). The immunotoxin was subsequently expressed in the tumour colonising oncolytic strain, Clostridium ghonii. Most importantly, the strictly anaerobic Clostridium ghonii was able to overexpress and secrete a functional CPE‑ETA' fusion protein. Our findings open the possibility of the targeted delivery of the immunotoxin locally to tumour sites at a high concentration using strictly anaerobic Clostridium ghonii for the treatment of CLDN4-positive cancer cells.
-
HER2-Specific Pseudomonas Exotoxin A PE25 Based Fusions: Influence of Targeting Domain on Target Binding, Toxicity, and In Vivo Biodistribution
Haozhong Ding,Mohamed Altai,Wen Yin,Sarah Lindbo,Hao Liu,Javad Garousi,Tianqi Xu,Anna Orlova,Vladimir Tolmachev,Sophia Hober,Torbjörn Gräslund
DOI: https://doi.org/10.3390/pharmaceutics12040391
IF: 6.525
2020-04-24
Pharmaceutics
Abstract:The human epidermal growth factor receptor 2 (HER2) is a clinically validated target for cancer therapy, and targeted therapies are often used in regimens for patients with a high HER2 expression level. Despite the success of current drugs, a number of patients succumb to their disease, which motivates development of novel drugs with other modes of action. We have previously shown that an albumin binding domain-derived affinity protein with specific affinity for HER2, ADAPT6, can be used to deliver the highly cytotoxic protein domain PE25, a derivative of Pseudomonas exotoxin A, to HER2 overexpressing malignant cells, leading to potent and specific cell killing. In this study we expanded the investigation for an optimal targeting domain and constructed two fusion toxins where a HER2-binding affibody molecule, ZHER2:2891, or the dual-HER2-binding hybrid ZHER2:2891-ADAPT6 were used for cancer cell targeting. We found that both targeting domains conferred strong binding to HER2; both to the purified extracellular domain and to the HER2 overexpressing cell line SKOV3. This resulted in fusion toxins with high cytotoxic potency toward cell lines with high expression levels of HER2, with EC50 values between 10 and 100 pM. For extension of the plasma half-life, an albumin binding domain was also included. Intravenous injection of the fusion toxins into mice showed a profound influence of the targeting domain on biodistribution. Compared to previous results, with ADAPT6 as targeting domain, ZHER2:2891 gave rise to further extension of the plasma half-life and also shifted the clearance route of the fusion toxin from the liver to the kidneys. Collectively, the results show that the targeting domain has a major impact on uptake of PE25-based fusion toxins in different organs. The results also show that PE25-based fusion toxins with high affinity to HER2 do not necessarily increase the cytotoxicity beyond a certain point in affinity. In conclusion, ZHER2:2891 has the most favorable characteristics as targeting domain for PE25.
-
The Progress in Immunoproapoptotin Molecules Targeting Tumor Cells
Tao Wang,Jun Ren,Lintao Jia,An‐Gang Yang
2013-01-01
Abstract:Immunoproapoptotin consists of a single-chain antibody against tumor-specific surface marker and an active domain which has the function of translocation and pro-apoptosis. This fused recombinant protein can specifically recognize and kill tumor cells. Immunoproapoptotin comes from immunotoxin which is proved not successful in clinical trials by inducing neutralizing antibody and other side effects due to the heterogenous toxins domains from bacteria or plants. To avoid the immunogenicity of immunotoxin, effectors from human were employed in the 2nd generation of Immunoproapoptotin. However, the translocation domain is still heterogenous and the efficiency of translocation is limited. In the latest progress, a furin cleavage site was used to substitute for the translocation domain and thus enhanced the translocation efficiency and greatly reduced the immunogenicity of Immunoproapoptotin, demonstrating a wide clinical application prospects.
-
Gigantoxin-4-4D5 scFv is a novel recombinant immunotoxin with specific toxicity against HER2/neu-positive ovarian carcinoma cells
Xinxin Lv,Jian Zhang,Rui Xu,Yuguo Dong,Aiyou Sun,Yaling Shen,Dongzhi Wei
DOI: https://doi.org/10.1007/s00253-016-7487-7
IF: 5
2016-01-01
Applied Microbiology and Biotechnology
Abstract:Immunotoxins are a new class of antibody-targeted therapy in clinical development. Traditional immunotoxins that are constructed from the toxins of plants or bacteria need to be internalized to the cytoplasm and thus have limited antitumor efficacy. In the present study, we combined a recently reported sea anemone cytolysin Gigantoxin-4 with an anti-HER2/neu single-chain variable fragment 4D5 scFv to construct a novel immunotoxin. We fused a SUMO tag to the N-terminus of Gigantoxin-4-4D5 scFv and it was successfully expressed in Escherichia coli strain BL21 (DE3) in a soluble form. After purification, the purity of Gigantoxin-4-4D5 scFv reached 96 % and the yield was 14.3 mg/L. Our results demonstrated that Gigantoxin-4-4D5 scFv exerted a highly cytotoxic effect on the HER2/neu-positive ovarian carcinoma SK-OV-3 cell line. And the hemolytic activity was weaker, making it safe for normal cells. The results of immunofluorescence analysis showed that this novel immunotoxin could specifically bind to SK-OV-3 cells with no recognition of human embryonic kidney 293 cells. Scanning electron microscope observations and extracellular lactate dehydrogenase activity indicated that it could induce necrosis in SK-OV-3 cells by disrupting the cell membrane. Moreover, it could also mediate apoptosis of SK-OV-3 cells.