Fig 1 - uploaded by Bernd Groner
Content may be subject to copyright.
Western blot analysis of Erb-B2 and EGF receptor expression in breast cancer cell lines, in CD34 blood progenitor cells, and in primary tumor cells isolated from breast cancer patients. Total protein was extracted and 100 g was run on a polyacrylamide gel unless otherwise indicated. After blotting, expression of Erb-B2 and EGF receptor was determined by staining with anti-Erb-B2 and anti-EGF receptor antisera as described in Materials and Methods.

Western blot analysis of Erb-B2 and EGF receptor expression in breast cancer cell lines, in CD34 blood progenitor cells, and in primary tumor cells isolated from breast cancer patients. Total protein was extracted and 100 g was run on a polyacrylamide gel unless otherwise indicated. After blotting, expression of Erb-B2 and EGF receptor was determined by staining with anti-Erb-B2 and anti-EGF receptor antisera as described in Materials and Methods.

Source publication
Article
Full-text available
Tumor cells have been found in autologous hematopoietic cell transplants used after high-dose chemotherapy. To specifically eliminate contaminating mammary tumor cells during ex vivo expansion of CD34+ hematopoietic progenitor cells, we used recombinant immunotoxins (ITs) directed against cell-surface antigens expressed on mammary carcinoma cells....

Citations

... Co-administration of cisplatin and scFv(14E1)-ETA was significantly more effective at killing the chemotherapy-resistant cells than either treatment alone. In preclinical studies of breast cancer patients, scFv(14E1)-ETA eliminated breast cancer cells from hematopoietic progenitor cell grafts ex vivo and was cytotoxic to 5/5 patient breast cancer tissue explants [143]. scFv(14E1)-ETA also demonstrated anti-metastatic capabilities, eliminating pulmonary metastases in an immunocompetent mouse model of metastatic renal cancer; however, all mice developed anti-PE antibodies resulting in neutralization of further toxin activity [144]. ...
Article
Full-text available
Many epithelial cancers rely on enhanced expression of the epidermal growth factor receptor (EGFR) to drive proliferation and survival pathways. Development of therapeutics to target EGFR signaling has been of high importance, and multiple examples have been approved for human use. However, many of the current small molecule or antibody-based therapeutics are of limited effectiveness due to the inevitable development of resistance and toxicity to normal tissues. Recombinant immunotoxins are therapeutic molecules consisting of an antibody or receptor ligand joined to a protein cytotoxin, combining the specific targeting of a cancer-expressed receptor with the potent cell killing of cytotoxic enzymes. Over the decades, many bacterial- or plant-based immunotoxins have been developed with the goal of targeting the broad range of cancers reliant upon EGFR overexpression. Many examples demonstrate excellent anti-cancer properties in preclinical development, and several EGFR-targeted immunotoxins have progressed to human trials. This review summarizes much of the past and current work in the development of immunotoxins for targeting EGFR-driven cancers.
... Proposed MM purging procedures must meet 2 important criteria: (1) they must effectively remove all contaminating cancer cells from the grafts and (2) they must fully spare the normal hematopoietic stem/progenitor cells (HSPCs) in the autograft allowing for successful reconstitution of the patient's hematopoietic system. Several purging methods have been explored in ASCT13141516, including a recent study focusing on ex vivo culture conditions that favor survival of HSPCs [17]. For MM, most of the focus has been placed on CD34 1 stem cell enrichment181920, which can reduce the level of MM contamination within the graft by 2 to 3 logs [20] . ...
Article
Full-text available
Autologous stem cell transplantation and novel therapies have improved overall survival of patients with multiple myeloma; however, most patients relapse and eventually succumb to their disease. Evidence indicates that residual cancer cells contaminate autologous grafts and may contribute to early relapses after autologous stem cell transplantation. Here, we demonstrate that ex vivo treatment with an oncolytic poxvirus called myxoma virus results in specific elimination of human myeloma cells by inducing rapid cellular apoptosis while fully sparing normal hematopoietic stem and progenitor cells. The specificity of this elimination is based on strong binding of the virus to myeloma cells coupled with an inability of the virus to bind or infect CD34(+) hematopoietic stem and progenitor cells. These 2 features allow myxoma to readily identify and distinguish even low levels of myeloma cells in complex mixtures. This ex vivo rabbit-specific oncolytic poxvirus called myxoma virus treatment also effectively inhibits systemic in vivo engraftment of human myeloma cells into immunodeficient mice and results in efficient elimination of primary CD138(+) myeloma cells contaminating patient hematopoietic cell products. We conclude that ex vivo myxoma treatment represents a safe and effective method to selectively eliminate myeloma cells from hematopoietic autografts before reinfusion.
... (i) chemotherapy with antiproliferative drugs such mafosfamide and 4-hydroperoxycyclophosphamide (4- HC, active metabolite of cyclophosphamide) [43, 44]; (ii) CD34+ stem/progenitor cell enrichment using immunomagnetic selection [45]; (iii) immunotoxins or hybrid cytotoxic proteins designed to selectively kill cancer cells such as heregulin (HRG) Pseudomonas exotoxin (PE) 40 [46]; (iv) immunomagnetic removal of tumor cells when the tumor cells express a unique antigen [47]; (v) monoclonal antibodies such as alemtuzumab (anti- CD52) and rituximab (anti-CD20) [48]; (vi) photodynamic purging by rhodamine [49]. ...
... Moreover, discriminating normal from malignant HSPC can be difficult when using just cell surface markers because the two populations are sometimes indistinguishable by immunophenotyping. Given similarities in immunophenotype , immunotoxins may target both malignant and normal HSPC, leading to impaired normal hematopoiesis and posttransplant hematopoietic reconstitution [46, 48]. Ideally, ex vivo purging is selective for the cancer cells yet spares normal HSPC. ...
Article
Full-text available
Hematological malignancies such as leukemias, lymphomas, multiple myeloma (MM), and the myelodysplastic syndromes (MDSs) primarily affect adults and are difficult to treat. For high-risk disease, hematopoietic stem cell transplant (HCT) can be used. However, in the setting of autologous HCT, relapse due to contamination of the autograft with cancer cells remains a major challenge. Ex vivo manipulations of the autograft to purge cancer cells using chemotherapies and toxins have been attempted. Because these past strategies lack specificity for malignant cells and often impair the normal hematopoietic stem and progenitor cells, prior efforts to ex vivo purge autografts have resulted in prolonged cytopenias and graft failure. The ideal ex vivo purging agent would selectively target the contaminating cancer cells while spare normal stem and progenitor cells and would be applied quickly without toxicities to the recipient. One agent which meets these criteria is oncolytic viruses. This paper details experimental progress with reovirus, myxoma virus, measles virus, vesicular stomatitis virus, coxsackievirus, and vaccinia virus as well as requirements for translation of these results to the clinic.
... Efficacy of scFv(FRP5)-ETA in the treatment of ErbB2-overex- pressing tumors has been established in numerous preclinical in vitro and in vivo studies. ScFv(FRP5)-ETA displayed potent antitumoral activity in vitro against a wide range of established and primary human tumor cells, including breast and ovarian carcinomas [9,14,15], squamous cell carcinomas [10,16] and prostate carcinomas [17]. In experimental animals, locally or systemically applied scFv(FRP5)-ETA effectively inhibited the in vivo growth of human tumor xenografts [9,10,14,16], and murine and rat tumor cells stably transfected with human c- erbB2 constructs [18,19]. ...
Article
Full-text available
ScFv(FRP5)-ETA is a recombinant antibody toxin with binding specificity for ErbB2 (HER2). It consists of an N-terminal single-chain antibody fragment (scFv), genetically linked to truncated Pseudomonas exotoxin A (ETA). Potent antitumoral activity of scFv(FRP5)-ETA against ErbB2-overexpressing tumor cells was previously demonstrated in vitro and in animal models. Here we report the first systemic application of scFv(FRP5)-ETA in human cancer patients. We have performed a phase I dose-finding study, with the objective to assess the maximum tolerated dose and the dose-limiting toxicity of intravenously injected scFv(FRP5)-ETA. Eighteen patients suffering from ErbB2-expressing metastatic breast cancers, prostate cancers, head and neck cancer, non small cell lung cancer, or transitional cell carcinoma were treated. Dose levels of 2, 4, 10, 12.5, and 20 microg/kg scFv(FRP5)-ETA were administered as five daily infusions each for two consecutive weeks. No hematologic, renal, and/or cardiovascular toxicities were noted in any of the patients treated. However, transient elevation of liver enzymes was observed, and considered dose limiting, in one of six patients at the maximum tolerated dose of 12.5 microg/kg, and in two of three patients at 20 microg/kg. Fifteen minutes after injection, peak concentrations of more than 100 ng/ml scFv(FRP5)-ETA were obtained at a dose of 10 microg/kg, indicating that predicted therapeutic levels of the recombinant protein can be applied without inducing toxic side effects. Induction of antibodies against scFv(FRP5)-ETA was observed 8 days after initiation of therapy in 13 patients investigated, but only in five of these patients could neutralizing activity be detected. Two patients showed stable disease and in three patients clinical signs of activity in terms of signs and symptoms were observed (all treated at doses > or = 10 microg/kg). Disease progression occurred in 11 of the patients. Our results demonstrate that systemic therapy with scFv(FRP5)-ETA can be safely administered up to a maximum tolerated dose of 12.5 microg/kg in patients with ErbB2-expressing tumors, justifying further clinical development.
... Efficacy of scFv(FRP5)-ETA in the treatment of ErbB2-overex- pressing tumors has been established in numerous preclinical in vitro and in vivo studies. ScFv(FRP5)-ETA displayed potent antitumoral activity in vitro against a wide range of established and primary human tumor cells, including breast and ovarian carcinomas [9,14,15], squamous cell carcinomas [10,16] and prostate carcinomas [17]. In experimental animals, locally or systemically applied scFv(FRP5)-ETA effectively inhibited the in vivo growth of human tumor xenografts [9,10,14,16], and murine and rat tumor cells stably transfected with human c- erbB2 constructs [18,19]. ...
Article
Full-text available
INTRODUCTION: ScFv(FRP5)-ETA is a recombinant antibody toxin with binding specificity for ErbB2 (HER2). It consists of an N-terminal single-chain antibody fragment (scFv), genetically linked to truncated Pseudomonas exotoxin A (ETA). Potent antitumoral activity of scFv(FRP5)-ETA against ErbB2-overexpressing tumor cells was previously demonstrated in vitro and in animal models. Here we report the first systemic application of scFv(FRP5)-ETA in human cancer patients. METHODS: We have performed a phase I dose-finding study, with the objective to assess the maximum tolerated dose and the dose-limiting toxicity of intravenously injected scFv(FRP5)-ETA. Eighteen patients suffering from ErbB2-expressing metastatic breast cancers, prostate cancers, head and neck cancer, non small cell lung cancer, or transitional cell carcinoma were treated. Dose levels of 2, 4, 10, 12.5, and 20 microg/kg scFv(FRP5)-ETA were administered as five daily infusions each for two consecutive weeks. RESULTS: No hematologic, renal, and/or cardiovascular toxicities were noted in any of the patients treated. However, transient elevation of liver enzymes was observed, and considered dose limiting, in one of six patients at the maximum tolerated dose of 12.5 microg/kg, and in two of three patients at 20 microg/kg. Fifteen minutes after injection, peak concentrations of more than 100 ng/ml scFv(FRP5)-ETA were obtained at a dose of 10 microg/kg, indicating that predicted therapeutic levels of the recombinant protein can be applied without inducing toxic side effects. Induction of antibodies against scFv(FRP5)-ETA was observed 8 days after initiation of therapy in 13 patients investigated, but only in five of these patients could neutralizing activity be detected. Two patients showed stable disease and in three patients clinical signs of activity in terms of signs and symptoms were observed (all treated at doses > or = 10 microg/kg). Disease progression occurred in 11 of the patients. CONCLUSION: Our results demonstrate that systemic therapy with scFv(FRP5)-ETA can be safely administered up to a maximum tolerated dose of 12.5 microg/kg in patients with ErbB2-expressing tumors, justifying further clinical development.
... In in vitro cell killing experiments scFv(FRP5)-ETA displayed potent antitumoral activity against a wide range of established and primary human tumor cells including breast and ovarian carcinomas [9,13,14], squamous cell carcinomas [15,16] and prostate carcinomas [17]. In experimental animals scFv(FRP5)-ETA effectively inhibited growth of established human tumor xenografts [9,13,15,16] and murine and rat tumor cells stably transfected with human c-erbB2 constructs [18][19][20]. ...
Article
Full-text available
ScFv(FRP5)-ETA is a recombinant single-chain antibody-toxin with binding specificity for ErbB2/HER2. Previously potent antitumoral activity of the molecule against ErbB2 overexpressing tumor cells was demonstrated in vitro and in animal models. Here we report on the first application of scFv(FRP5)-ETA in human cancer patients summarizing case reports collected in four different clinical centers. Eleven patients suffering from metastatic breast and colorectal cancers and from malignant melanoma were treated on a compassionate-use basis by intratumoral injection of scFv(FRP5)-ETA into cutaneous lesions once daily for 7-10 days. Total daily doses ranged from 60 to 900 microg, and total doses per treatment cycle ranged from 0.6 to 6.0 mg. Treatment caused injected tumors to shrink in six of the 10 cases evaluated (60%). Complete regression of injected tumor nodules was accomplished in four patients (40%) and partial reduction in tumor size in another two patients (20%). Adverse reactions were restricted to local symptoms such as pain and inflammation at injection sites which were fully reversible. Only in one patient treated at the highest daily doses systemic liver toxicity of grade 2 was observed and treatment was discontinued on day 7. No hematologic, renal, and/or cardiovascular toxicities were noted. Our results demonstrate that local therapy with scFv(FRP5)-ETA can be effective against ErbB2 expressing tumors justifying further clinical development of this reagent.
... This is supported by the findings of Vredenburgh et al (1997), who reported that the higher the number of tumour cells identified in the bone marrow harvest, the shorter the disease-free and overall survival. A variety of techniques have been proposed to purge tumour cells ex vivo (Gribben et al, 1991; Vogel et al, 1996; Spyridonidis et al, 1998). Although prospective randomized trials remain to be investigated to determine whether a positive selection of haematopoietic progenitor cells and/or depletion of tumour cells contaminating PBSC fraction affects clinical outcome, the combination of in vivo and ex vivo purging might become an attractive strategy to improve patient survival. ...
Article
Full-text available
High-dose chemotherapy with peripheral blood stem cell (PBSC) transplantation in advanced germ cell tumour (GCT) patients is widely applied. The aims of this study were: (1) To examine the presence of alphafetoprotein (AFP) bearing tumour cells in PBSC harvests from advanced GCT patients obtained after multiple cycles of induction chemotherapy. (2) To determine whether induction chemotherapy contributed to in vivo purging of the tumour. We evaluated cryopreserved PBSC samples from 5 patients with advanced stage II/III AFP producing GCT. PBSC were separated after the first, second and third cycles of induction chemotherapy. Those samples were analysed using the nested reverse transcription polymerase chain reaction (RT-PCR) method to detect AFP mRNA. Although, in all patients, AFP mRNA was detected in PBSC samples after the first or second cycle of induction chemotherapy, but was not detected in 3 of 4 samples after the third cycle of chemotherapy. Although it is not clear whether tumour cells contaminating PBSC fraction contribute to disease relapse, PBSC harvested after at least 3 cycles of induction chemotherapy might be recommended to avoid such a possibility.
... Whereas hematopoietic growth factors do not influence survival and proliferation of primary breast cancer cells, we found a strong tumor-cell suppressive effect when using serum-free preparations or when TGF-pi was added to the cultures [70]. This technology may be combined with other purging techniques, like molecular depletion by immunotoxins [71], creating highly efficient purging strategies. Assessment of the clinical value of ex vivo tumor depletion methods awaits the results of clinical trials. ...
Article
Full-text available
The increasing use of high-dose chemotherapy with autologous hematopoietic transplantation for the treatment of solid malignancies has raised concern about the role of tumor cells contaminating the grafts. Minimal residual disease (MRD) in autologous grafts has became a dynamic and intensively studied field in oncology. This review discusses the current status of MRD in breast cancer autografts and presents existing data on detection methodology, clinical relevance, biologic characteristics and purging techniques.
Article
Malignant cell contamination in autologous transplants is a potential origin of tumor relapse. Ex vivo expansion of CD34+ blood progenitor cells (BPC) has been proposed as a tool to eliminate tumor cells from autografts. To characterize the influence of culture conditions on survival, growth, and clonogenicity of malignant cells, we isolated primary mammary carcinoma cells from pleural effusions and ascites of patients with metastatic breast cancer and cultured them in the presence of stem cell factor (SCF), interleukin-1β (IL-1β), IL-3, IL-6, and erythropoietin (EPO), ie, conditions previously shown to allow efficient ex vivo expansion of CD34+ BPC. In the presence of serum, tumor cells proliferated during a 7-day culture period and no significant growth-modulatory effect was attributable to the presence of hematopoietic growth factors. When transforming growth factor-β1 (TGF-β1) was added to these cultures, proliferation of breast cancer cells was reduced. Expansion of clonogenic tumor cells was seen in the presence of SCF + IL-1β + IL-3 + IL-6 + EPO, but was suppressed by TGF-β1. Cocultures of tumor cells in direct cellular contact with hematopoietic cells showed that tumor cell growth could be stimulated by ex vivo expanded hematopoietic cells at high cell densities (5 × 105/mL). In contrast, culture under serum-free conditions resulted in death of greater than 90% of breast cancer cells within 7 days and a further decrease in tumor cell numbers thereafter. In the serum-free cultures, hematopoietic cytokines and cellular contact with CD34+ BPC could not protect the tumor cells from death. Therefore, ex vivo expansion of CD34+ BPC in serum-free medium provides an environment for efficient purging of contaminating mammary carcinoma cells. These results have clinical significance for future protocols in autologous progenitor cell transplantation in cancer patients.
Article
The continuously growing natural killer (NK) cell line NK-92 is highly cytotoxic against malignant cells of various origins without affecting normal human cells. Based on this selectivity, the potential of NK-92 cells for adoptive therapy is currently being investigated in phase I clinical studies. To further enhance the antitumoral activity of NK-92 cells and expand the range of tumor entities suitable for NK-92–based therapies, here by transduction with a retroviral vector we have generated genetically modified NK-92 cells expressing a chimeric antigen receptor specific for the tumor-associated ErbB2 (HER2/neu) antigen, which is overexpressed by many tumors of epithelial origin. The chimeric antigen receptor consists of the ErbB2-specific scFv(FRP5) antibody fragment, a flexible hinge region derived from CD8, and transmembrane and intracellular regions of the CD3 ζ chain. Transduced NK-92-scFv(FRP5)-ζ cells express high levels of the fusion protein on the cell surface as determined by fluorescence-activated cell-scanning (FACS) analysis. In europium release assays, no difference in cytotoxic activity of NK-92 and NK-92-scFv(FRP5)-ζ cells toward ErbB2-negative targets was found. However, even at low effector-to-target ratios, NK-92-scFv(FRP5)-ζ cells specifically and efficiently lysed established and primary ErbB2-expressing tumor cells that were completely resistant to cytolytic activity of parental NK-92 cells. These results demonstrate that efficient retargeting of NK-92 cytotoxicity can be achieved and might allow the generation of potent cell-based therapeutics for the treatment of ErbB2-expressing malignancies.