50 results on '"Boghaert ER"'
Search Results
2. BCL-X L -targeting antibody-drug conjugates are active in preclinical models and mitigate on-mechanism toxicity of small-molecule inhibitors.
- Author
-
Judd AS, Bawa B, Buck WR, Tao ZF, Li Y, Mitten MJ, Bruncko M, Catron N, Doherty G, Durbin KR, Enright B, Frey R, Haasch D, Haman S, Haight AR, Henriques TA, Holms J, Izeradjene K, Judge RA, Jenkins GJ, Kunzer A, Leverson JD, Martin RL, Mitra D, Mittelstadt S, Nelson L, Nimmer P, Palma J, Peterson R, Phillips DC, Ralston SL, Rosenberg SH, Shen X, Song X, Vaidya KS, Wang X, Wang J, Xiao Y, Zhang H, Zhang X, Blomme EA, Boghaert ER, Kalvass JC, Phillips A, and Souers AJ
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Small Molecule Libraries pharmacology, bcl-X Protein antagonists & inhibitors, bcl-X Protein metabolism, Immunoconjugates pharmacology, Xenograft Model Antitumor Assays
- Abstract
Overexpression of the antiapoptotic protein B-cell lymphoma-extra large (BCL-X
L ) is associated with drug resistance and disease progression in numerous cancers. The compelling nature of this protein as a therapeutic target prompted efforts to develop selective small-molecule BCL-XL inhibitors. Although efficacious in preclinical models, we report herein that selective BCL-XL inhibitors cause severe mechanism-based cardiovascular toxicity in higher preclinical species. To overcome this liability, antibody-drug conjugates were constructed using altered BCL-XL -targeting warheads, unique linker technologies, and therapeutic antibodies. The epidermal growth factor receptor-targeting antibody-drug conjugate AM1-15 inhibited growth of tumor xenografts and did not cause cardiovascular toxicity nor dose-limiting thrombocytopenia in monkeys. While an unprecedented BCL-XL -mediated toxicity was uncovered in monkey kidneys upon repeat dosing of AM1-15, this toxicity was mitigated via further drug-linker modification to afford AM1-AAA (AM1-25). The AAA drug-linker has since been incorporated into mirzotamab clezutoclax, the first selective BCL-XL -targeting agent to enter human clinical trials.- Published
- 2024
- Full Text
- View/download PDF
3. Tackling the tumor microenvironment - how can complex tumor models in vitro aid oncology drug development?
- Author
-
Cox MC, Mendes R, Halwachs KN, Domenici G, Brito C, and Boghaert ER
- Subjects
- Humans, Tumor Microenvironment, Drug Delivery Systems methods, Drug Development, Neoplasms pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
Introduction: Identifying effective cancer drugs remains an inefficient process. Drug efficacy in traditional preclinical cancer models translates poorly into therapy in the clinic. Implementation of preclinical models that incorporate the tumor microenvironment (TME) is needed to improve selection of active drugs prior to clinical trials., Areas Covered: Progression of cancer results from the behavior of cancer cells in concert with the host's histopathological background. Nonetheless, complex preclinical models with a relevant microenvironment have yet to become an integral part of drug development. This review discusses existing models and provides a synopsis of active areas of cancer drug development where implementation would be of value. Their contribution to finding therapeutics in immune oncology, angiogenesis, regulated cell death and targeting tumor fibroblasts as well as optimization of drug delivery, combination therapy, and biomarkers of efficacy is considered., Expert Opinion: Complex tumor models in vitro (CTMIVs) that mimic the organotypic architecture of neoplastic tumors have boosted research into TME influence on traditional cytoreductive chemotherapy as well as the detection of specific TME targets. Despite advances in technical prowess, CTMIVs can only address specific aspects of cancer pathophysiology.
- Published
- 2023
- Full Text
- View/download PDF
4. Exploring Metabolic Signatures of Ex Vivo Tumor Tissue Cultures for Prediction of Chemosensitivity in Ovarian Cancer.
- Author
-
Mendes R, Graça G, Silva F, Guerreiro ACL, Gomes-Alves P, Serpa J, Boghaert ER, Alves PM, Félix A, Brito C, and Isidro IA
- Abstract
Predicting patient response to treatment and the onset of chemoresistance are still major challenges in oncology. Chemoresistance is deeply influenced by the complex cellular interactions occurring within the tumor microenvironment (TME), including metabolic crosstalk. We have previously shown that ex vivo tumor tissue cultures derived from ovarian carcinoma (OvC) resections retain the TME components for at least four weeks of culture and implemented assays for assessment of drug response. Here, we explored ex vivo patient-derived tumor tissue cultures to uncover metabolic signatures of chemosensitivity and/or resistance. Tissue cultures derived from nine OvC cases were challenged with carboplatin and paclitaxel, the standard-of-care chemotherapeutics, and the metabolic footprints were characterized by LC-MS. Partial least-squares discriminant analysis (PLS-DA) revealed metabolic signatures that discriminated high-responder from low-responder tissue cultures to ex vivo drug exposure. As a proof-of-concept, a set of potential metabolic biomarkers of drug response was identified based on the receiver operating characteristics (ROC) curve, comprising amino acids, fatty acids, pyrimidine, glutathione, and TCA cycle pathways. Overall, this work establishes an analytical and computational platform to explore metabolic features of the TME associated with response to treatment, which can leverage the discovery of biomarkers of drug response and resistance in OvC.
- Published
- 2022
- Full Text
- View/download PDF
5. Pathophysiologic and Pharmacologic Considerations to Improve the Design and Application of Antibody-Drug Conjugates.
- Author
-
Boghaert ER, Cox MC, and Vaidya KS
- Subjects
- Antibodies therapeutic use, Humans, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Immunoconjugates chemistry, Immunoconjugates pharmacology, Immunoconjugates therapeutic use, Neoplasms drug therapy
- Abstract
Antibody-drug conjugates (ADC) have emerged as one of the pillars of clinical disease management in oncology. The biggest hurdle to widespread development and application of ADCs has been a narrow therapeutic index. Advances in antibody technologies and formats as well as novel linker and payload chemistries have begun to facilitate structural improvements to ADCs. However, the interplay of structural characteristics with physiologic and pharmacologic factors determining therapeutic success has garnered less attention. This review elaborates on the pharmacology of ADCs, the pathophysiology of cancerous tissues, and the reciprocal consequences on ADC properties and functions. While most currently approved ADCs utilize either microtubule inhibition or DNA damage as primary mechanisms of action, we present arguments to expand this repertoire and highlight the need for payload mechanisms that exploit disease-specific vulnerabilities. We promote the idea that the choice of antibody format, targeting antigen, linker properties, and payload of an ADC should be deliberately fit for purpose by taking the pathophysiology of disease and the specific pharmacology of the drug entity into account, thus allowing a higher probability of clinical success., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
6. Pevonedistat and azacitidine upregulate NOXA (PMAIP1) to increase sensitivity to venetoclax in preclinical models of acute myeloid leukemia.
- Author
-
Cojocari D, Smith BN, Purkal JJ, Arrate MP, Huska JD, Xiao Y, Gorska A, Hogdal LJ, Ramsey HE, Boghaert ER, Phillips DC, and Savona MR
- Subjects
- Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cyclopentanes, Humans, Pyrimidines, Sulfonamides, Azacitidine pharmacology, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology
- Abstract
Dysregulation of apoptotic machinery is one mechanism by which acute myeloid leukemia (AML) acquires a clonal survival advantage. B-cell lymphoma protein-2 (BCL2) overexpression is a common feature in hematologic malignancies. The selective BCL2 inhibitor, venetoclax (VEN) is used in combination with azacitidine (AZA), a DNAmethyltransferase inhibitor (DNMTi), to treat patients with AML. Despite promising response rates to VEN/AZA, resistance to the agent is common. One identified mechanism of resistance is the upregulation of myeloid cell leukemia-1 protein (MCL1). Pevonedistat (PEV), a novel agent that inhibits NEDD8-activating enzyme, and AZA both upregulate NOXA (PMAIP1), a BCL2 family protein that competes with effector molecules at the BH3 binding site of MCL1. We demonstrate that PEV/AZA combination induces NOXA to a greater degree than either PEV or AZA alone, which enhances VEN-mediated apoptosis. Herein, using AML cell lines and primary AML patient samples ex vivo, including in cells with genetic alterations linked to treatment resistance, we demonstrate robust activity of the PEV/VEN/AZA triplet. These findings were corroborated in preclinical systemic engrafted models of AML. Collectively, these results provide rational for combining PEV/VEN/AZA as a novel therapeutic approach in overcoming AML resistance in current therapies.
- Published
- 2022
- Full Text
- View/download PDF
7. Patient-Derived Explants of Colorectal Cancer: Histopathological and Molecular Analysis of Long-Term Cultures.
- Author
-
da Mata S, Franchi-Mendes T, Abreu S, Filipe B, Morgado S, Mesquita M, Albuquerque C, Fonseca R, Santo VE, Boghaert ER, Rosa I, and Brito C
- Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. Although short-term cultures of tumour sections and xenotransplants have been used to determine drug efficacy, the results frequently fail to confer clinically useful information. Biomarker discovery has changed the paradigm for advanced CRC, though the presence of a biomarker does not necessarily translate into therapeutic success. To improve clinical outcomes, translational models predictive of drug response are needed. We describe a simple method for the fast establishment of CRC patient-derived explant (CRC-PDE) cultures from different carcinogenesis pathways, employing agitation-based platforms. A total of 26 CRC-PDE were established and a subset was evaluated for viability ( n = 23), morphology and genetic key alterations ( n = 21). CRC-PDE retained partial tumor glandular architecture and microenvironment features were partially lost over 4 weeks of culture. Key proteins (p53 and Mismatch repair) and oncogenic driver mutations of the original tumours were sustained throughout the culture. Drug challenge ( n = 5) revealed differential drug response from distinct CRC-PDE cases. These findings suggest an adequate representation of the original tumour and highlight the importance of detailed model characterisation. The preservation of key aspects of the CRC microenvironment and genetics supports CRC-PDE potential applicability in pre- and co-clinical settings, as long as temporal dynamics are considered.
- Published
- 2021
- Full Text
- View/download PDF
8. Application of LDH assay for therapeutic efficacy evaluation of ex vivo tumor models.
- Author
-
Cox MC, Mendes R, Silva F, Mendes TF, Zelaya-Lazo A, Halwachs K, Purkal JJ, Isidro IA, Félix A, Boghaert ER, and Brito C
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Drug Discovery methods, Humans, Mice, Spheroids, Cellular drug effects, Spheroids, Cellular metabolism, Tumor Cells, Cultured, Drug Screening Assays, Antitumor methods, L-Lactate Dehydrogenase metabolism, Neoplasms drug therapy, Neoplasms metabolism
- Abstract
The current standard preclinical oncology models are not able to fully recapitulate therapeutic targets and clinically relevant disease biology, evidenced by the 90% attrition rate of new therapies in clinical trials. Three-dimensional (3D) culture systems have the potential to enhance the relevance of preclinical models. However, the limitations of currently available cellular assays to accurately evaluate therapeutic efficacy in these models are hindering their widespread adoption. We assessed the compatibility of the lactate dehydrogenase (LDH) assay in 3D spheroid cultures against other commercially available readout methods. We developed a standardized protocol to apply the LDH assay to ex vivo cultures, considering the impact of culture growth dynamics. We show that accounting for growth rates and background release levels of LDH are sufficient to make the LDH assay a suitable methodology for longitudinal monitoring and endpoint assessment of therapeutic efficacy in both cell line-derived xenografts (xenospheres) and patient-derived explant cultures. This method has the added value of being non-destructive and not dependent on reagent penetration or manipulation of the parent material. The establishment of reliable readout methods for complex 3D culture systems will further the utility of these tumor models in preclinical and co-clinical drug development studies., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
9. Synergistic therapeutic benefit by combining the antibody drug conjugate, depatux-m with temozolomide in pre-clinical models of glioblastoma with overexpression of EGFR.
- Author
-
Vaidya KS, Mitten MJ, Zelaya-Lazo AL, Oleksijew A, Alvey C, Falls HD, Mishra S, Palma J, Ansell P, Phillips AC, Reilly EB, Anderson M, and Boghaert ER
- Subjects
- Animals, Cell Proliferation drug effects, Disease Models, Animal, Drug Synergism, ErbB Receptors genetics, Humans, Mice, Xenograft Model Antitumor Assays, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Brain Neoplasms genetics, Brain Neoplasms pathology, Glioblastoma genetics, Glioblastoma pathology, Temozolomide pharmacology
- Abstract
Purpose: Depatux-m is an antibody drug conjugate (ADC) that targets and inhibits growth of cancer cells overexpressing the epidermal growth factor receptor (EGFR) or the 2-7 deletion mutant (EGFRvIII) in tumor models in vitro and in vivo. Treatment of patients suffering from relapsed/refractory glioblastoma (GBM) with a combination of depatux-m and temozolomide (TMZ) tended to increase overall survival. As a first step to understand the nature of the interaction between the two drugs, we investigated whether the interaction was synergistic, additive or antagonistic., Methods: The efficacy of ADCs, antibodies, TMZ and radiation was tested in xenograft models of GBM, U-87MG and U-87MG EGFRvIII. Both models express EGFR. U-87MG EGFRvIII was transduced to express EGFRvIII. Changes in tumor volume, biomarkers of cell death and apoptosis after treatment were used to measure efficacy of the various treatments. Synergism of depatux-m and TMZ was verified in three-dimensional cultures of U-87MG and U-87MG EGFRvIII by the method of Chou and Talalay., Results: Combined with TMZ and radiotherapy (RT), depatux-m inhibited xenograft growth of U-87MG and U-87MG EGFRvIII more than either treatment with depatux-m or TMZ + RT. Durability of the response to depatux-m + TMZ + RT or depatux-m + TMZ was more pronounced in U-87MG EGFRvIII than in U-87MG. Efficacy of depatux-m + TMZ was synergistic in U-87MG EGFRvIII and additive in U-87MG., Conclusion: Adding depatux-m enhances the efficacy of standard of care therapy in preclinical models of GBM. Durability of response to depatux-m + TMZ in vivo and synergy of the drug-drug interaction correlates with the amount of antigen expressed by the tumor cells.
- Published
- 2021
- Full Text
- View/download PDF
10. Patient-derived ovarian cancer explants: preserved viability and histopathological features in long-term agitation-based cultures.
- Author
-
Abreu S, Silva F, Mendes R, Mendes TF, Teixeira M, Santo VE, Boghaert ER, Félix A, and Brito C
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents pharmacology, Cell Survival drug effects, Epithelial Cells metabolism, Female, Fibroblasts metabolism, Humans, Ki-67 Antigen analysis, Lymphocytes, Tumor-Infiltrating metabolism, Middle Aged, Ovarian Neoplasms metabolism, Time Factors, Tumor Cells, Cultured, Cell Culture Techniques methods, Epithelial Cells pathology, Fibroblasts pathology, Lymphocytes, Tumor-Infiltrating pathology, Ovarian Neoplasms pathology
- Abstract
Ovarian carcinoma (OvC) remains a major therapeutic challenge due to its propensity to develop resistance after an initial response to chemotherapy. Interactions of tumour cells with the surrounding microenvironment play a role in tumour survival, invasion capacity and drug resistance. Cancer models that retain tissue architecture and tumour microenvironment components are therefore essential to understand drug response and resistance mechanisms. Herein, our goal was to develop a long-term OvC patient-derived explant (OvC-PDE) culture strategy in which architecture and cell type heterogeneity of the original tumour would be retained. Samples from 25 patients with distinct OvC types and one with a benign tumour, were cultured for 30 days in agitation-based culture systems with 100% success rate. OvC-PDE cultures retained the original tumour architecture and main cellular components: epithelial cells, fibroblasts and immune cells. Epithelial cells kept their original levels of proliferation and apoptosis. Moreover, the major extracellular components, such as collagen-I and -IV, were retained in explants. OvC-PDE cultures were exposed to standard-of-care chemotherapeutics agents for 2 weeks, attesting the ability of the platform for drug assays employing cyclic drug exposure regimens. We established an OvC-PDE dynamic culture in which tumour architecture and cell type heterogeneity were preserved for the different OvC types, replicating features of the original tumour and compatible with long-term drug exposure for drug efficacy and resistance studies.
- Published
- 2020
- Full Text
- View/download PDF
11. Targeting Multiple EGFR-expressing Tumors with a Highly Potent Tumor-selective Antibody-Drug Conjugate.
- Author
-
Anderson MG, Falls HD, Mitten MJ, Oleksijew A, Vaidya KS, Boghaert ER, Gao W, Palma JP, Cao D, Chia PL, John T, Gan HK, Scott AM, and Reilly EB
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Immunoconjugates pharmacology, Mice, Mice, Nude, ErbB Receptors metabolism, Immunoconjugates therapeutic use
- Abstract
ABBV-321 (serclutamab talirine), a next-generation EGFR-targeted antibody-drug conjugate (ADC) incorporates a potent pyrrolobenzodiazepine (PBD) dimer toxin conjugated to the EGFR-targeting ABT-806 affinity-matured AM1 antibody. ABBV-321 follows the development of related EGFR-targeted ADCs including depatuxizumab mafodotin (depatux-m, ABT-414), ABT-806 conjugated to monomethyl auristatin F (MMAF), and ABBV-221 (losatuxizumab vedotin), AM1 antibody conjugated to monomethyl auristatin E (MMAE). The distinct tumor selectivity of ABBV-321 differentiates it from many previous highly active antibody PBD conjugates that lack a therapeutic window. Potency of the PBD dimer, combined with increased binding of AM1 to EGFR-positive tumor cells, opens the possibility to target a wide array of tumors beyond those with high levels of EGFR overexpression or amplification, including those insensitive to auristatin-based ADCs. ABBV-321 exhibits potent antitumor activity in cellular and in vivo studies including xenograft cell line and patient-derived xenograft glioblastoma, colorectal, lung, head and neck, and malignant mesothelioma tumor models that are less sensitive to depatux-m or ABBV-221. Combination studies with ABBV-321 and depatux-m suggest a promising treatment option permitting suboptimal, and potentially better tolerated, doses of both ADCs while providing improved potency. Collectively, these data suggest that ABBV-321 may offer an extended breadth of efficacy relative to other EGFR ADCs while extending utility to multiple EGFR-expressing tumor indications. Despite its highly potent PBD dimer payload, the tumor selectivity of ABBV-321, coupled with its pharmacology, toxicology, and pharmacokinetic profiles, support continuation of ongoing phase I clinical trials in patients with advanced EGFR-expressing malignancies., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
12. 5-Azacitidine Induces NOXA to Prime AML Cells for Venetoclax-Mediated Apoptosis.
- Author
-
Jin S, Cojocari D, Purkal JJ, Popovic R, Talaty NN, Xiao Y, Solomon LR, Boghaert ER, Leverson JD, and Phillips DC
- Subjects
- Animals, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Cell Line, Tumor, DNA Methylation, Disease Models, Animal, Dose-Response Relationship, Drug, Drug Synergism, Gene Expression Regulation, Leukemic drug effects, Gene Knockdown Techniques, Humans, Leukemia, Myeloid, Acute, Mice, Proto-Oncogene Proteins c-bcl-2 metabolism, Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis genetics, Azacitidine pharmacology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Proto-Oncogene Proteins c-bcl-2 genetics, Sulfonamides pharmacology
- Abstract
Purpose: Patients with acute myeloid leukemia (AML) frequently do not respond to conventional therapies. Leukemic cell survival and treatment resistance have been attributed to the overexpression of B-cell lymphoma 2 (BCL-2) and aberrant DNA hypermethylation. In a phase Ib study in elderly patients with AML, combining the BCL-2 selective inhibitor venetoclax with hypomethylating agents 5-azacitidine (5-Aza) or decitabine resulted in 67% overall response rate; however, the underlying mechanism for this activity is unknown., Experimental Design: We studied the consequences of combining two therapeutic agents, venetoclax and 5-Aza, in AML preclinical models and primary patient samples. We measured expression changes in the integrated stress response (ISR) and the BCL-2 family by Western blot and qPCR. Subsequently, we engineered PMAIP1 (NOXA)- and BBC3 (PUMA)-deficient AML cell lines using CRISPR- Cas9 methods to understand their respective roles in driving the venetoclax/5-Aza combinatorial activity., Results: In this study, we demonstrate that venetoclax and 5-Aza act synergistically to kill AML cells in vitro and display combinatorial antitumor activity in vivo . We uncover a novel nonepigenetic mechanism for 5-Aza-induced apoptosis in AML cells through transcriptional induction of the proapoptotic BH3-only protein NOXA. This induction occurred within hours of treatment and was mediated by the ISR pathway. NOXA was detected in complex with antiapoptotic proteins, suggesting that 5-Aza may be "priming" the AML cells for venetoclax-induced apoptosis. PMAIP1 knockout confirmed its major role in driving venetoclax and 5-Aza synergy., Conclusions: These data provide a novel nonepigenetic mechanism of action for 5-Aza and its combinatorial activity with venetoclax through the ISR-mediated induction of PMAIP1 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
13. Discovery of A-1331852, a First-in-Class, Potent, and Orally-Bioavailable BCL-X L Inhibitor.
- Author
-
Wang L, Doherty GA, Judd AS, Tao ZF, Hansen TM, Frey RR, Song X, Bruncko M, Kunzer AR, Wang X, Wendt MD, Flygare JA, Catron ND, Judge RA, Park CH, Shekhar S, Phillips DC, Nimmer P, Smith ML, Tahir SK, Xiao Y, Xue J, Zhang H, Le PN, Mitten MJ, Boghaert ER, Gao W, Kovar P, Choo EF, Diaz D, Fairbrother WJ, Elmore SW, Sampath D, Leverson JD, and Souers AJ
- Abstract
Herein we describe the discovery of A-1331852, a first-in-class orally active BCL-X
L inhibitor that selectively and potently induces apoptosis in BCL-XL -dependent tumor cells. This molecule was generated by re-engineering our previously reported BCL-XL inhibitor A-1155463 using structure-based drug design. Key design elements included rigidification of the A-1155463 pharmacophore and introduction of sp3 -rich moieties capable of generating highly productive interactions within the key P4 pocket of BCL-XL . A-1331852 has since been used as a critical tool molecule for further exploring BCL-2 family protein biology, while also representing an attractive entry into a drug discovery program., Competing Interests: The authors declare the following competing financial interest(s): AbbVie and Genentech participated in interpretation of data and review and approval of publication. L.W., G.A.D., A.S.J., Z-F.T., T. M. H., R.R.F., X.S., M.B., A.R.K., X.W., M.D.W., N.D.C., S.S., D.C.P., R.A.J., P.N., M.L.S., S.K.T., Y.X., J.X., H.Z., P.N.L., M.J.M., E.R.B., W.G., P.K., S.W.E., J.D.L., and A.J.S. are employees of AbbVie, Inc. C.H.P. was an employee of AbbVie, Inc at the time of the study. E.C. and W.J.F. are employees of Genentech, Inc. J.A.F., D.D., and D.S. were employees of Genentech, Inc. at the time of the study.- Published
- 2020
- Full Text
- View/download PDF
14. Characterization of ABBV-221, a Tumor-Selective EGFR-Targeting Antibody Drug Conjugate.
- Author
-
Phillips AC, Boghaert ER, Vaidya KS, Falls HD, Mitten MJ, DeVries PJ, Benatuil L, Hsieh CM, Meulbroek JA, Panchal SC, Buchanan FG, Durbin KR, Voorbach MJ, Reuter DR, Mudd SR, Loberg LI, Ralston SL, Cao D, Gan HK, Scott AM, and Reilly EB
- Subjects
- Animals, Antibodies, Monoclonal, Humanized chemistry, Apoptosis, Cell Proliferation, ErbB Receptors antagonists & inhibitors, ErbB Receptors immunology, Female, Glioblastoma metabolism, Glioblastoma pathology, Humans, Immunoconjugates chemistry, Mice, Mice, Inbred BALB C, Mice, Nude, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antibodies, Monoclonal, Humanized pharmacology, Glioblastoma drug therapy, Immunoconjugates pharmacology, Oligopeptides chemistry
- Abstract
Depatuxizumab mafodotin (depatux-m, ABT-414) is a tumor-selective antibody drug conjugate (ADC) comprised of the anti-EGFR antibody ABT-806 and the monomethyl auristatin F (MMAF) warhead. Depatux-m has demonstrated promising clinical activity in glioblastoma multiforme (GBM) patients and is currently being evaluated in clinical trials in first-line and recurrent GBM disease settings. Depatux-m responses have been restricted to patients with amplified EGFR, highlighting the need for therapies with activity against tumors with nonamplified EGFR overexpression. In addition, depatux-m dosing has been limited by corneal side effects common to MMAF conjugates. We hypothesized that a monomethyl auristatin E (MMAE) ADC utilizing an EGFR-targeting antibody with increased affinity may have broader utility against tumors with more modest EGFR overexpression while mitigating the risk of corneal side effects. We describe here preclinical characterization of ABBV-221, an EGFR-targeting ADC comprised of an affinity-matured ABT-806 conjugated to MMAE. ABBV-221 binds to a similar EGFR epitope as depatux-m and retains tumor selectivity with increased binding to EGFR-positive tumor cells and greater in vitro potency. ABBV-221 displays increased tumor uptake and antitumor activity against wild-type EGFR-positive xenografts with a greatly reduced incidence of corneal side effects relative to depatux-m. ABBV-221 has similar activity as depatux-m against an EGFR-amplified GBM patient derived xenograft (PDX) model and is highly effective alone and in combination with standard-of-care temozolomide in an EGFRvIII-positive GBM xenograft model. Based on these results, ABBV-221 has advanced to a phase I clinical trial in patients with advanced solid tumors associated with elevated levels of EGFR. Mol Cancer Ther; 17(4); 795-805. ©2018 AACR., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
15. The Volume of Three-Dimensional Cultures of Cancer Cells InVitro Influences Transcriptional Profile Differences and Similarities with Monolayer Cultures and Xenografted Tumors.
- Author
-
Boghaert ER, Lu X, Hessler PE, McGonigal TP, Oleksijew A, Mitten MJ, Foster-Duke K, Hickson JA, Santo VE, Brito C, Uziel T, and Vaidya KS
- Subjects
- Animals, Cell Culture Techniques, Cell Line, Tumor, Cluster Analysis, Disease Models, Animal, Female, Gene Expression Profiling, Heterografts, Humans, Mice, Spheroids, Cellular, Gene Expression Regulation, Neoplastic, Transcriptome
- Abstract
Improving the congruity of preclinical models with cancer as it is manifested in humans is a potential way to mitigate the high attrition rate of new cancer therapies in the clinic. In this regard, three-dimensional (3D) tumor cultures in vitro have recently regained interest as they have been acclaimed to have higher similarity to tumors in vivo than to cells grown in monolayers (2D). To identify cancer functions that are active in 3D rather than in 2D cultures, we compared the transcriptional profiles (TPs) of two non-small cell lung carcinoma cell lines, NCI-H1650 and EBC-1 grown in both conditions to the TP of xenografted tumors. Because confluence, diameter or volume can hypothetically alter TPs, we made intra- and inter-culture comparisons using samples with defined dimensions. As projected by Ingenuity Pathway Analysis (IPA), a limited number of signal transduction pathways operational in vivo were better represented by 3D than by 2D cultures in vitro. Growth of 2D and 3D cultures as well as xenografts induced major changes in the TPs of these 3 modes of culturing. Alterations of transcriptional network activation that were predicted to evolve similarly during progression of 3D cultures and xenografts involved the following functions: hypoxia, proliferation, cell cycle progression, angiogenesis, cell adhesion, and interleukin activation. Direct comparison of TPs of 3D cultures and xenografts to monolayer cultures yielded up-regulation of networks involved in hypoxia, TGF and Wnt signaling as well as regulation of epithelial mesenchymal transition. Differences in TP of 2D and 3D cancer cell cultures are subject to progression of the cultures. The emulation of the predicted cell functions in vivo is therefore not only determined by the type of culture in vitro but also by the confluence or diameter of the 2D or 3D cultures, respectively. Consequently, the successful implementation of 3D models will require phenotypic characterization to verify the relevance of applying these models for drug development., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
16. ABBV-399, a c-Met Antibody-Drug Conjugate that Targets Both MET -Amplified and c-Met-Overexpressing Tumors, Irrespective of MET Pathway Dependence.
- Author
-
Wang J, Anderson MG, Oleksijew A, Vaidya KS, Boghaert ER, Tucker L, Zhang Q, Han EK, Palma JP, Naumovski L, and Reilly EB
- Subjects
- Animals, Antibodies, Monoclonal adverse effects, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Mice, Neoplasms immunology, Neoplasms pathology, Proto-Oncogene Proteins c-met antagonists & inhibitors, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Antibodies, Monoclonal administration & dosage, Neoplasms drug therapy, Neoplasms genetics, Proto-Oncogene Proteins c-met genetics
- Abstract
Purpose: Despite the importance of the MET oncogene in many malignancies, clinical strategies targeting c-Met have benefitted only small subsets of patients with tumors driven by signaling through the c-Met pathway, thereby necessitating selection of patients with MET amplification and/or c-Met activation most likely to respond. An ADC targeting c-Met could overcome these limitations with potential as a broad-acting therapeutic. Experimental Design: ADC ABBV-399 was generated with the c-Met-targeting antibody, ABT-700. Antitumor activity was evaluated in cancer cells with overexpressed c-Met or amplified MET and in xenografts including patient-derived xenograft (PDX) models and those refractory to other c-Met inhibitors. The correlation between c-Met expression and sensitivity to ABBV-399 in tumor and normal cell lines was assessed to evaluate the risk of on-target toxicity. Results: A threshold level of c-Met expressed by sensitive tumor but not normal cells is required for significant ABBV-399-mediated killing of tumor cells. Activity extends to c-Met or amplified MET cell line and PDX models where significant tumor growth inhibition and regressions are observed. ABBV-399 inhibits growth of xenograft tumors refractory to other c-Met inhibitors and provides significant therapeutic benefit in combination with standard-of-care chemotherapy. Conclusions: ABBV-399 represents a novel therapeutic strategy to deliver a potent cytotoxin to c-Met-overexpressing tumor cells enabling cell killing regardless of reliance on MET signaling. ABBV-399 has progressed to a phase I study where it has been well tolerated and has produced objective responses in c-Met-expressing non-small cell lung cancer (NSCLC) patients. Clin Cancer Res; 23(4); 992-1000. ©2016 AACR ., (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
17. A "Prozone-Like" Effect Influences the Efficacy of the Monoclonal Antibody ABT-700 against the Hepatocyte Growth Factor Receptor.
- Author
-
Vaidya KS, Oleksijew A, Tucker LA, Pappano WN, Anderson MG, Grinnell CM, Zhang Q, Heighton SJ, Mitten MJ, Mishra S, Palma JP, Wang J, Reilly EB, and Boghaert ER
- Subjects
- Animals, Cell Line, Cisplatin administration & dosage, Humans, Mice, Mice, Nude, Mice, SCID, Antibodies, Monoclonal pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Hepatocytes drug effects, Hepatocytes metabolism, Proto-Oncogene Proteins c-met metabolism
- Abstract
ABT-700 is a therapeutic antibody against the hepatocyte growth factor receptor (MET). At doses or regimens that lead to exposures exceeding optimum in vivo, the efficacy of ABT-700 is unexpectedly reduced. We hypothesized that this reduction in efficacy was due to a "prozone-like" effect in vivo. A prozone-like effect, which is a reduction in efficacy beyond optimum exposure, is caused due a mechanism similar to the generation of false negative flocculation tests by excessive antibody titres. In vitro, we demonstrate that at higher ABT-700 concentrations, this "prozone-like" effect is mediated by a progressive conversion from bivalent to ineffective monovalent binding of the antibody. In vivo, the efficacy of ABT-700 is dependent on an optimum range of exposure as well. Our data suggest that the "prozone-like" effect is operative and independent of target expression. ABT-700 dose, regimen, exposure, and tumor burden are interdependent variables influencing the "prozone-like" effect and mediating and in vivo efficacy. By optimization of dosage and regimen we demonstrate that the "prozone-like" effect can be alleviated and ABT-700 efficacy at varying tumor loads can be further extended in combination with cisplatin. Our results suggest that optimization of exposure taking tumor burden into account may alleviate "prozone-like" effects without compromising efficacy., (© 2017 S. Karger AG, Basel.)
- Published
- 2017
- Full Text
- View/download PDF
18. Expression Profile of BCL-2, BCL-XL, and MCL-1 Predicts Pharmacological Response to the BCL-2 Selective Antagonist Venetoclax in Multiple Myeloma Models.
- Author
-
Punnoose EA, Leverson JD, Peale F, Boghaert ER, Belmont LD, Tan N, Young A, Mitten M, Ingalla E, Darbonne WC, Oleksijew A, Tapang P, Yue P, Oeh J, Lee L, Maiga S, Fairbrother WJ, Amiot M, Souers AJ, and Sampath D
- Subjects
- Animals, Bcl-2-Like Protein 11 metabolism, Bortezomib pharmacology, Cell Line, Tumor, Disease Models, Animal, Drug Resistance, Neoplasm genetics, Drug Therapy, Combination, Humans, Immunohistochemistry, Mice, Multiple Myeloma drug therapy, Multiple Myeloma metabolism, Protein Binding, Proto-Oncogene Proteins c-bcl-2 metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Gene Expression Regulation, Neoplastic, Multiple Myeloma genetics, Myeloid Cell Leukemia Sequence 1 Protein genetics, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 genetics, Sulfonamides pharmacology, bcl-X Protein genetics
- Abstract
BCL-2 family proteins dictate survival of human multiple myeloma cells, making them attractive drug targets. Indeed, multiple myeloma cells are sensitive to antagonists that selectively target prosurvival proteins such as BCL-2/BCL-XL (ABT-737 and ABT-263/navitoclax) or BCL-2 only (ABT-199/GDC-0199/venetoclax). Resistance to these three drugs is mediated by expression of MCL-1. However, given the selectivity profile of venetoclax it is unclear whether coexpression of BCL-XL also affects antitumor responses to venetoclax in multiple myeloma. In multiple myeloma cell lines (n = 21), BCL-2 is expressed but sensitivity to venetoclax correlated with high BCL-2 and low BCL-XL or MCL-1 expression. Multiple myeloma cells that coexpress BCL-2 and BCL-XL were resistant to venetoclax but sensitive to a BCL-XL-selective inhibitor (A-1155463). Multiple myeloma xenograft models that coexpressed BCL-XL or MCL-1 with BCL-2 were also resistant to venetoclax. Resistance to venetoclax was mitigated by cotreatment with bortezomib in xenografts that coexpressed BCL-2 and MCL-1 due to upregulation of NOXA, a proapoptotic factor that neutralizes MCL-1. In contrast, xenografts that expressed BCL-XL, MCL-1, and BCL-2 were more sensitive to the combination of bortezomib with a BCL-XL selective inhibitor (A-1331852) but not with venetoclax cotreatment when compared with monotherapies. IHC of multiple myeloma patient bone marrow biopsies and aspirates (n = 95) revealed high levels of BCL-2 and BCL-XL in 62% and 43% of evaluable samples, respectively, while 34% were characterized as BCL-2(High)/BCL-XL (Low) In addition to MCL-1, our data suggest that BCL-XL may also be a potential resistance factor to venetoclax monotherapy and in combination with bortezomib. Mol Cancer Ther; 15(5); 1132-44. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
19. ABT-414, an Antibody-Drug Conjugate Targeting a Tumor-Selective EGFR Epitope.
- Author
-
Phillips AC, Boghaert ER, Vaidya KS, Mitten MJ, Norvell S, Falls HD, DeVries PJ, Cheng D, Meulbroek JA, Buchanan FG, McKay LM, Goodwin NC, and Reilly EB
- Subjects
- Animals, Antibody Affinity, Cell Line, Tumor, Cell Survival drug effects, Combined Modality Therapy, Disease Models, Animal, ErbB Receptors genetics, ErbB Receptors immunology, ErbB Receptors metabolism, Female, Glioblastoma drug therapy, Glioblastoma metabolism, Glioblastoma mortality, Glioblastoma pathology, Humans, Molecular Targeted Therapy, Mutation, Protein Binding, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Epitopes immunology, ErbB Receptors antagonists & inhibitors, Immunoconjugates pharmacology, Protein Kinase Inhibitors pharmacology
- Abstract
Targeting tumor-overexpressed EGFR with an antibody-drug conjugate (ADC) is an attractive therapeutic strategy; however, normal tissue expression represents a significant toxicity risk. The anti-EGFR antibody ABT-806 targets a unique tumor-specific epitope and exhibits minimal reactivity to EGFR in normal tissue, suggesting its suitability for the development of an ADC. We describe the binding properties and preclinical activity of ABT-414, an ABT-806 monomethyl auristatin F conjugate. In vitro, ABT-414 selectively kills tumor cells overexpressing wild-type or mutant forms of EGFR. ABT-414 inhibits the growth of xenograft tumors with high EGFR expression and causes complete regressions and cures in the most sensitive models. Tumor growth inhibition is also observed in tumor models with EGFR mutations, including activating mutations and those with the exon 2-7 deletion [EGFR variant III (EGFRvIII)], commonly found in glioblastoma multiforme. ABT-414 exhibits potent cytotoxicity against glioblastoma multiforme patient-derived xenograft models expressing either wild-type EGFR or EGFRvIII, with sustained regressions and cures observed at clinically relevant doses. ABT-414 also combines with standard-of-care treatment of radiation and temozolomide, providing significant therapeutic benefit in a glioblastoma multiforme xenograft model. On the basis of these results, ABT-414 has advanced to phase I/II clinical trials, and objective responses have been observed in patients with both amplified wild-type and EGFRvIII-expressing tumors. Mol Cancer Ther; 15(4); 661-9. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
20. Clearance of systemic hematologic tumors by venetoclax (Abt-199) and navitoclax.
- Author
-
Ackler S, Oleksijew A, Chen J, Chyla BJ, Clarin J, Foster K, McGonigal T, Mishra S, Schlessinger S, Smith ML, Tahir SK, Leverson JD, Souers AJ, Boghaert ER, and Hickson J
- Abstract
The Bcl-2 family inhibitors venetoclax and navitoclax demonstrated potent antitumor activity in chronic lymphocytic leukemia patients, notably in reducing marrow load and adenopathy. Subsequent trials with venetoclax have been initiated in non-Hodgkin's lymphoma and multiple myeloma patients. Traditional preclinical models fall short either in faithfully recapitulating disease progression within such compartments or in allowing the direct longitudinal analysis of systemic disease. We show that intravenous inoculation of engineered RS4;11 (acute lymphoblastic leukemia) and Granta 519 (mantle cell lymphoma) bioluminescent reporter cell lines result in tumor engraftment of bone marrow, with additional invasion of the central nervous system in the case of Granta 519. Importantly, apoptosis induction and response of these systemically engrafted tumors to Bcl-2 family inhibitors alone or in combination with standard-of-care agents could be monitored longitudinally with optical imaging, and was more accurately reflective of the observed clinical response.
- Published
- 2015
- Full Text
- View/download PDF
21. Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy.
- Author
-
Leverson JD, Phillips DC, Mitten MJ, Boghaert ER, Diaz D, Tahir SK, Belmont LD, Nimmer P, Xiao Y, Ma XM, Lowes KN, Kovar P, Chen J, Jin S, Smith M, Xue J, Zhang H, Oleksijew A, Magoc TJ, Vaidya KS, Albert DH, Tarrant JM, La N, Wang L, Tao ZF, Wendt MD, Sampath D, Rosenberg SH, Tse C, Huang DC, Fairbrother WJ, Elmore SW, and Souers AJ
- Subjects
- Administration, Oral, Aniline Compounds therapeutic use, Animals, Antineoplastic Agents therapeutic use, Benzothiazoles chemistry, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cell Line, Tumor, Cell Survival, Docetaxel, Gene Expression Profiling, Granulocytes metabolism, Humans, Isoquinolines chemistry, Kinetics, Mice, Neoplasm Transplantation, Neoplasms metabolism, Neutropenia chemically induced, Neutrophils drug effects, Proto-Oncogene Proteins c-bcl-2 metabolism, Sulfonamides therapeutic use, Taxoids adverse effects, Thrombocytopenia chemically induced, bcl-X Protein antagonists & inhibitors, bcl-X Protein metabolism, Gene Expression Regulation, Neoplastic, Neoplasms drug therapy, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors
- Abstract
The BCL-2/BCL-XL/BCL-W inhibitor ABT-263 (navitoclax) has shown promising clinical activity in lymphoid malignancies such as chronic lymphocytic leukemia. However, its efficacy in these settings is limited by thrombocytopenia caused by BCL-XL inhibition. This prompted the generation of the BCL-2-selective inhibitor venetoclax (ABT-199/GDC-0199), which demonstrates robust activity in these cancers but spares platelets. Navitoclax has also been shown to enhance the efficacy of docetaxel in preclinical models of solid tumors, but clinical use of this combination has been limited by neutropenia. We used venetoclax and the BCL-XL-selective inhibitors A-1155463 and A-1331852 to assess the relative contributions of inhibiting BCL-2 or BCL-XL to the efficacy and toxicity of the navitoclax-docetaxel combination. Selective BCL-2 inhibition suppressed granulopoiesis in vitro and in vivo, potentially accounting for the exacerbated neutropenia observed when navitoclax was combined with docetaxel clinically. By contrast, selectively inhibiting BCL-XL did not suppress granulopoiesis but was highly efficacious in combination with docetaxel when tested against a range of solid tumors. Therefore, BCL-XL-selective inhibitors have the potential to enhance the efficacy of docetaxel in solid tumors and avoid the exacerbation of neutropenia observed with navitoclax. These studies demonstrate the translational utility of this toolkit of selective BCL-2 family inhibitors and highlight their potential as improved cancer therapeutics., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
22. Discovery of a Potent and Selective BCL-XL Inhibitor with in Vivo Activity.
- Author
-
Tao ZF, Hasvold L, Wang L, Wang X, Petros AM, Park CH, Boghaert ER, Catron ND, Chen J, Colman PM, Czabotar PE, Deshayes K, Fairbrother WJ, Flygare JA, Hymowitz SG, Jin S, Judge RA, Koehler MF, Kovar PJ, Lessene G, Mitten MJ, Ndubaku CO, Nimmer P, Purkey HE, Oleksijew A, Phillips DC, Sleebs BE, Smith BJ, Smith ML, Tahir SK, Watson KG, Xiao Y, Xue J, Zhang H, Zobel K, Rosenberg SH, Tse C, Leverson JD, Elmore SW, and Souers AJ
- Abstract
A-1155463, a highly potent and selective BCL-XL inhibitor, was discovered through nuclear magnetic resonance (NMR) fragment screening and structure-based design. This compound is substantially more potent against BCL-XL-dependent cell lines relative to our recently reported inhibitor, WEHI-539, while possessing none of its inherent pharmaceutical liabilities. A-1155463 caused a mechanism-based and reversible thrombocytopenia in mice and inhibited H146 small cell lung cancer xenograft tumor growth in vivo following multiple doses. A-1155463 thus represents an excellent tool molecule for studying BCL-XL biology as well as a productive lead structure for further optimization.
- Published
- 2014
- Full Text
- View/download PDF
23. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets.
- Author
-
Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J, Dayton BD, Ding H, Enschede SH, Fairbrother WJ, Huang DC, Hymowitz SG, Jin S, Khaw SL, Kovar PJ, Lam LT, Lee J, Maecker HL, Marsh KC, Mason KD, Mitten MJ, Nimmer PM, Oleksijew A, Park CH, Park CM, Phillips DC, Roberts AW, Sampath D, Seymour JF, Smith ML, Sullivan GM, Tahir SK, Tse C, Wendt MD, Xiao Y, Xue JC, Zhang H, Humerickhouse RA, Rosenberg SH, and Elmore SW
- Subjects
- Aniline Compounds pharmacology, Animals, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Cell Survival drug effects, Dogs, Female, HeLa Cells, Humans, Mice, Mice, SCID, Proto-Oncogene Proteins c-bcl-2 chemistry, Tumor Burden, Xenograft Model Antitumor Assays, bcl-X Protein antagonists & inhibitors, Antineoplastic Agents pharmacology, Blood Platelets drug effects, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Hematologic Neoplasms drug therapy, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Sulfonamides pharmacology
- Abstract
Proteins in the B cell CLL/lymphoma 2 (BCL-2) family are key regulators of the apoptotic process. This family comprises proapoptotic and prosurvival proteins, and shifting the balance toward the latter is an established mechanism whereby cancer cells evade apoptosis. The therapeutic potential of directly inhibiting prosurvival proteins was unveiled with the development of navitoclax, a selective inhibitor of both BCL-2 and BCL-2-like 1 (BCL-X(L)), which has shown clinical efficacy in some BCL-2-dependent hematological cancers. However, concomitant on-target thrombocytopenia caused by BCL-X(L) inhibition limits the efficacy achievable with this agent. Here we report the re-engineering of navitoclax to create a highly potent, orally bioavailable and BCL-2-selective inhibitor, ABT-199. This compound inhibits the growth of BCL-2-dependent tumors in vivo and spares human platelets. A single dose of ABT-199 in three patients with refractory chronic lymphocytic leukemia resulted in tumor lysis within 24 h. These data indicate that selective pharmacological inhibition of BCL-2 shows promise for the treatment of BCL-2-dependent hematological cancers.
- Published
- 2013
- Full Text
- View/download PDF
24. Delineation of a cellular hierarchy in lung cancer reveals an oncofetal antigen expressed on tumor-initiating cells.
- Author
-
Damelin M, Geles KG, Follettie MT, Yuan P, Baxter M, Golas J, DiJoseph JF, Karnoub M, Huang S, Diesl V, Behrens C, Choe SE, Rios C, Gruzas J, Sridharan L, Dougher M, Kunz A, Hamann PR, Evans D, Armellino D, Khandke K, Marquette K, Tchistiakova L, Boghaert ER, Abraham RT, Wistuba II, and Zhou BB
- Subjects
- Animals, CD24 Antigen analysis, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Epithelial-Mesenchymal Transition, Humans, Hyaluronan Receptors analysis, Lung Neoplasms immunology, Lung Neoplasms pathology, Membrane Glycoproteins physiology, Mice, Antigens, Neoplasm analysis, Carcinoma, Non-Small-Cell Lung drug therapy, Immunotoxins therapeutic use, Lung Neoplasms drug therapy, Membrane Glycoproteins analysis, Neoplastic Stem Cells immunology
- Abstract
Poorly differentiated tumors in non-small cell lung cancer (NSCLC) have been associated with shorter patient survival and shorter time to recurrence following treatment. Here, we integrate multiple experimental models with clinicopathologic analysis of patient tumors to delineate a cellular hierarchy in NSCLC. We show that the oncofetal protein 5T4 is expressed on tumor-initiating cells and associated with worse clinical outcome in NSCLC. Coexpression of 5T4 and factors involved in the epithelial-to-mesenchymal transition were observed in undifferentiated but not in differentiated tumor cells. Despite heterogeneous expression of 5T4 in NSCLC patient-derived xenografts, treatment with an anti-5T4 antibody-drug conjugate resulted in complete and sustained tumor regression. Thus, the aggressive growth of heterogeneous solid tumors can be blocked by therapeutic agents that target a subpopulation of cells near the top of the cellular hierarchy.
- Published
- 2011
- Full Text
- View/download PDF
25. Determination of pharmacokinetic values of calicheamicin-antibody conjugates in mice by plasmon resonance analysis of small (5 microl) blood samples.
- Author
-
Boghaert ER, Khandke KM, Sridharan L, Dougher M, DiJoseph JF, Kunz A, Hamann PR, Moran J, Chaudhary I, and Damle NK
- Subjects
- Aminoglycosides administration & dosage, Aminoglycosides blood, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal blood, Antibodies, Monoclonal, Humanized, Area Under Curve, Cell Line, Tumor, Gemtuzumab, Half-Life, Humans, Injections, Intraperitoneal, Injections, Intravenous, Inotuzumab Ozogamicin, Mice, Mice, Inbred BALB C, Mice, Nude, Rabbits, Surface Plasmon Resonance, Aminoglycosides pharmacokinetics, Antibodies, Monoclonal pharmacokinetics
- Abstract
Purpose: The present study aims to establish a method that provides fast, precise and reproducible pharmacokinetic (PK) parameters of antibody-calicheamicin conjugates. The method should discriminate between PK of the antibody moiety and PK of the conjugated calicheamicin (CM)., Methods: The conjugates gemtuzumab ozogamicin (CMA-676, Mylotarg) or inotuzumab ozogamicin (CMC-544) were injected in the tail vein of nude mice. At regular time intervals, 5 mul whole blood samples were taken from the tail artery. Concentrations of conjugated CMA-676 or CMC-544 as well as concentrations of their respective antibody moiety were determined by sandwich plasmon resonance. This detection system measures changes in the plasma resonance angle caused by the interaction of macromolecules on biosensor chips. We determined as a first measure the binding of CMA-676 or CMC-544 to their respective antigens, CD33 or CD22. As a second measure we determined the amount of CM on the antigen-bound conjugates. This was done by determination of changes in plasma resonance angle after binding of an anti-CM antibody., Results: Sandwich plasmon resonance allowed detection of both conjugates in blood of mice in a range of 100-1,000 ng/ml protein. Due to the precision of the sampling and detection methods, PK values of each conjugate were determined in individual mice. Calicheamicin bound to antibody was eliminated faster than the antibody alone. The presence of a CD22-expressing tumour in mice reduced the plasma levels of the CD22-targeting conjugate but not of the CD33-targeting one., Conclusions: Using small blood samples from a mouse, the sandwich plasmon resonance method provided PK-values of CM-conjugates and information about the stability of the linkage in vivo. Comparison between the PK-values of CM-conjugates in tumour-bearing and tumour-free mice suggested that retention of the conjugate in tumour tissue due to antigen targeting could be deduced from the plasma levels.
- Published
- 2008
- Full Text
- View/download PDF
26. The oncofetal protein, 5T4, is a suitable target for antibody-guided anti-cancer chemotherapy with calicheamicin.
- Author
-
Boghaert ER, Sridharan L, Khandke KM, Armellino D, Ryan MG, Myers K, Harrop R, Kunz A, Hamann PR, Marquette K, Dougher M, DiJoseph JF, and Damle NK
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma pathology, Animals, Antibodies, Monoclonal, Humanized, Antigens, Neoplasm analysis, Antigens, Neoplasm genetics, Antigens, Neoplasm physiology, Cell Line, Tumor, Female, Gemtuzumab, Humans, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Mice, Mice, Nude, Neoplasm Transplantation, Transplantation, Heterologous, Aminoglycosides therapeutic use, Antibodies, Monoclonal therapeutic use, Antigens, Neoplasm immunology, Antineoplastic Agents therapeutic use
- Abstract
The oncofetal protein, 5T4, is a tumor-associated protein displayed on the cell membrane of various carcinomas. This molecule is a promising target for anti-tumor vaccine development and for targeted therapy with staphylococcus exotoxin. The potential use of 5T4 as a target for antibody-guided chemotherapy has not been demonstrated. We report oncolytic efficacy and selectivity in vitro and in vivo with immuno-conjugates of calicheamicin (CM) and the anti-5T4 antibody, H8. CM is a potent cytotoxic drug that causes double strand breaks in DNA. Conjugates of CM and H8 were constructed with acid-labile as well as acid-stabile linkers. In vitro, when applied to monolayers of 5T4(+) cells, CM-conjugates targeting 5T4 were consistently more toxic than either free drug or a non-binding control CM-conjugate. This difference was less pronounced on 5T4-deficient cells. In vivo, four 5T4-positive subcutaneous tumor models were treated with conjugates. Efficacy was demonstrated by reduction of tumor growth relative to controls treated with drug vehicle. To evidence selectivity, the efficacy of the anti-5T4 conjugates was compared to the efficacy of H8, a mixture of H8 and calicheamicin, calicheamicin alone or calicheamicin conjugated to the anti-CD33 antibody, hP67.6. In addition, the efficacy and selectivity of an acid-labile conjugate of H8 was evaluated in an orthotopic model for 5T4(+) lung cancer. Increased survival following treatment was used as a parameter of efficacy. Calicheamicin conjugates of H8 were effective and selective in all the examined tumor models. Differences in efficacy between the acid-labile and acid-stabile conjugates depended on the investigated tumor model.
- Published
- 2008
- Full Text
- View/download PDF
27. CD20-specific antibody-targeted chemotherapy of non-Hodgkin's B-cell lymphoma using calicheamicin-conjugated rituximab.
- Author
-
Dijoseph JF, Dougher MM, Armellino DC, Kalyandrug L, Kunz A, Boghaert ER, Hamann PR, and Damle NK
- Subjects
- Aminoglycosides immunology, Animals, Antibiotics, Antineoplastic administration & dosage, Antibiotics, Antineoplastic immunology, Antibodies, Monoclonal immunology, Antibodies, Monoclonal, Murine-Derived, Antibody Specificity, Cell Proliferation drug effects, Cytotoxicity, Immunologic drug effects, Enediynes immunology, Flow Cytometry, Humans, Immunoconjugates immunology, Lymphoma, B-Cell immunology, Mice, Mice, Nude, Mice, SCID, Rituximab, Aminoglycosides administration & dosage, Antibodies, Monoclonal administration & dosage, Antigens, CD20 immunology, Drug Delivery Systems methods, Enediynes administration & dosage, Immunoconjugates administration & dosage, Lymphoma, B-Cell drug therapy
- Abstract
Tumor-targeted delivery of a potent cytotoxic agent, calicheamicin, using its immunoconjugates is a clinically validated therapeutic strategy. Rituximab is a human CD20-specific chimeric antibody extensively used in B-NHL therapy. We investigated whether conjugation to calicheamicin can improve the anti-tumor activity of rituximab against human B-cell lymphoma (BCL) xenografts in preclinical models. BCL cells were cultured with rituximab or its calicheamicin conjugates and their in vitro growth was monitored. BCL cells were injected s.c. to establish localized xenografts in nude mice or i.v. to establish disseminated BCL in severe combined immunodeficient (scid) mice. I.p. treatment with rituximab or its calicheamicin conjugates was initiated and its effect on s.c. BCL growth or survival of mice with disseminated BCL was monitored. Conjugation of calicheamicin to rituximab vastly enhanced its growth inhibitory activity against BCL in vitro. Conjugation to calicheamicin had no deleterious effect on the effector functional activity of rituximab. Calicheamicin conjugated to rituximab with an acid-labile linker exhibited greater anti-tumor activity against s.c. BCL xenografts and improved survival of mice with disseminated BCL over that of unconjugated rituximab. Anti-tumor activities of rituximab conjugated to calicheamicin via an acid-stable linker were similar to that of unconjugated rituximab. Superior anti-tumor efficacy exhibited by a calicheamicin immunoconjugate of rituximab with an acid-labile linker over that of rituximab demonstrates the therapeutic potential of CD20-specific antibody-targeted chemotherapy strategy in the treatment of B-NHL.
- Published
- 2007
- Full Text
- View/download PDF
28. Tumoricidal effect of calicheamicin immuno-conjugates using a passive targeting strategy.
- Author
-
Boghaert ER, Khandke K, Sridharan L, Armellino D, Dougher M, Dijoseph JF, Kunz A, Hamann PR, Sridharan A, Jones S, Discafani C, and Damle NK
- Subjects
- Aminoglycosides pharmacokinetics, Aminoglycosides pharmacology, Animals, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Antibodies, Monoclonal, Murine-Derived, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Cell Line, Tumor, Cell Survival drug effects, Female, Gemtuzumab, HT29 Cells, Humans, Immunoconjugates pharmacokinetics, Immunoconjugates pharmacology, Immunoglobulin Fc Fragments chemistry, Immunoglobulin Fc Fragments therapeutic use, Inhibitory Concentration 50, Male, Mice, Mice, Nude, Polyethylene Glycols chemistry, Rituximab, Serum Albumin therapeutic use, Aminoglycosides therapeutic use, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Immunoconjugates therapeutic use, Xenograft Model Antitumor Assays methods
- Abstract
Calicheamicin is a potent chemotherapeutic with a low therapeutic index that requires targeting to tumor cells for its use in the clinic. To treat acute myeloid leukemia, calicheamicin has been conjugated to an antibody that recognizes CD33 (gemtuzumab ozogamicin). The application range of this 'active' targeting strategy is limited since it depends on specific antigen expression by tumor cells. This limitation could be reduced by using an antigen-independent 'passive targeting' strategy for calicheamicin. 'Passive targeting' relies on the dysfunctional vasculature of a neoplastic tumor that allows enhanced retention of macromolecules. We studied the efficacy of calicheamicin conjugated to various carrier molecules: i.e. immunoglobulin, albumin or PEGylated Fc fragments. In nude mice, a conjugate of anti-CD33 and calicheamicin accumulates in human tumor xenografts in the absence of detectable amounts of targeting antigen. Passive targeting provided sufficient accumulation of this conjugate to inhibit tumor growth of 10 different CD33-negative xenograft models. This efficacy depended on the use of an acid-labile linker between antibody and calicheamicin. Substitution of immunoglobulin as a carrier with either albumin or PEGylated Fc reduced or eliminated the efficacy of the conjugate. The results showed that using 'non-specific' immunoglobulin for passive targeting of calicheamicin might be an effective mode of cancer therapy.
- Published
- 2006
29. Antitumor efficacy of a combination of CMC-544 (inotuzumab ozogamicin), a CD22-targeted cytotoxic immunoconjugate of calicheamicin, and rituximab against non-Hodgkin's B-cell lymphoma.
- Author
-
DiJoseph JF, Dougher MM, Kalyandrug LB, Armellino DC, Boghaert ER, Hamann PR, Moran JK, and Damle NK
- Subjects
- Aminoglycosides chemistry, Aminoglycosides immunology, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal, Humanized, Antibodies, Monoclonal, Murine-Derived, Cell Line, Tumor, Cytotoxicity, Immunologic drug effects, Female, Flow Cytometry, Humans, Immunologic Factors administration & dosage, Inotuzumab Ozogamicin, Male, Mice, Mice, Nude, Mice, SCID, Rituximab, Sialic Acid Binding Ig-like Lectin 2 drug effects, Sialic Acid Binding Ig-like Lectin 2 immunology, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Immunoconjugates pharmacology, Lymphoma, B-Cell drug therapy, Neoplasms, Experimental drug therapy
- Abstract
Purpose: CMC-544 is a CD22-targeted cytotoxic immunoconjugate, currently being evaluated in B-cell non-Hodgkin's lymphoma (B-NHL) patients. Rituximab is a CD20-targeted antibody commonly used in B-NHL therapy. Here, we describe antitumor efficacy of a combination of CMC-544 and rituximab against B-cell lymphoma (BCL) in preclinical models., Experimental Design: BCLs were cultured in vitro with CMC-544, rituximab, or their combination. BCLs were injected either s.c. or i.v. to establish localized s.c. BCL in nude mice or disseminated BCL in severe combined immunodeficient mice, respectively. I.p. treatment with CMC-544 or rituximab was initiated at various times either alone or in combination and its effect on s.c. BCL growth or survival of mice with disseminated BCL was monitored., Results: In vitro growth-inhibitory activity of CMC-544 combined with rituximab was additive. Rituximab but not CMC-544 exhibited effector functions, such as antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. Rituximab was less effective in inhibiting growth of established BCL xenografts than developing xenografts. In contrast, CMC-544 was equally effective against both developing and established BCL xenografts. Although CMC-544 and rituximab individually caused partial inhibition of the growth of BCL xenografts at suboptimal doses examined, their combination suppressed xenograft growth by >90%. In a disseminated BCL model, 60% of CMC-544-treated mice and 20% of rituximab-treated mice survived for 125 days. In contrast, 90% of mice treated with the combination of CMC-544 and rituximab survived for longer than 125 days., Conclusion: The demonstration of superior antitumor activity of a combination of CMC-544 and rituximab described here provides the preclinical basis for its clinical evaluation as a treatment option for B-NHL.
- Published
- 2006
- Full Text
- View/download PDF
30. Antibody-targeted chemotherapy of B-cell lymphoma using calicheamicin conjugated to murine or humanized antibody against CD22.
- Author
-
DiJoseph JF, Popplewell A, Tickle S, Ladyman H, Lawson A, Kunz A, Khandke K, Armellino DC, Boghaert ER, Hamann P, Zinkewich-Peotti K, Stephens S, Weir N, and Damle NK
- Subjects
- Amino Acid Sequence, Aminoglycosides chemistry, Aminoglycosides immunology, Animals, Antibodies, Monoclonal genetics, Antibodies, Monoclonal immunology, Antineoplastic Agents immunology, Binding, Competitive, Cell Line, Tumor, Epitopes immunology, Female, Humans, Immunoconjugates immunology, Lymphoma, B-Cell immunology, Mice, Mice, Inbred BALB C, Mice, Nude, Molecular Sequence Data, Sialic Acid Binding Ig-like Lectin 2, Xenograft Model Antitumor Assays methods, Aminoglycosides therapeutic use, Antibodies, Monoclonal therapeutic use, Antigens, CD immunology, Antigens, Differentiation, B-Lymphocyte immunology, Antineoplastic Agents therapeutic use, Cell Adhesion Molecules immunology, Immunoconjugates therapeutic use, Lectins immunology, Lymphoma, B-Cell therapy
- Abstract
Antibody-targeted chemotherapy with immunoconjugates of calicheamicin is a clinically validated strategy in cancer therapy. This study describes the selection of a murine anti-CD22 mAb, m5/44, as a targeting agent, its conjugation to a derivative of calicheamicin (CalichDM) via either acid-labile or acid-stable linkers, the antitumor activity of CalichDM conjugated to m5/44, and its subsequent humanization by CDR grafting. Murine IgG1 mAb m5/44 was selected based on its subnanomolar affinity for CD22 and ability to be internalized into B cells. CalichDM conjugated to m5/44 caused potent growth inhibition of CD22+ human B-cell lymphomas (BCLs) in vitro. The conjugate of m5/44 with an acid-labile linker was more potent than an acid-stable conjugate, a nonbinding conjugate with a similar acid-labile linker, or unconjugated CalichDMH in inhibiting BCL growth. CalichDM conjugated to m5/44 caused regression of established BCL xenografts in nude mice. In contrast, both unconjugated m5/44 and a nonbinding conjugate were ineffective against these xenografts. Based on the potent antitumor activity of m5/44-CalichDM conjugates, m5/44 was humanized by CDR grafting to create g5/44, an IgG4 anti-CD22 antibody. Both m5/44 and g5/44 bound CD22 with subnanomolar affinity. Competitive blocking with previously characterized murine anti-CD22 mAbs suggested that g5/44 recognizes epitope A located within the first N-terminal Ig-like domain of human CD22. Antitumor efficacy of CalichDM conjugated to g5/44 against BCL xenografts was more potent than its murine counterpart. Based on these results, a calicheamicin conjugate of g5/44, CMC-544, was selected for further development as a targeted chemotherapeutic agent for the treatment of B-cell malignancies.
- Published
- 2005
- Full Text
- View/download PDF
31. Potent and specific antitumor efficacy of CMC-544, a CD22-targeted immunoconjugate of calicheamicin, against systemically disseminated B-cell lymphoma.
- Author
-
DiJoseph JF, Goad ME, Dougher MM, Boghaert ER, Kunz A, Hamann PR, and Damle NK
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Antibodies, Monoclonal, Murine-Derived, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Humans, Immunoglobulin G metabolism, Immunotherapy methods, Inotuzumab Ozogamicin, Lymphoma, B-Cell metabolism, Lymphoma, B-Cell pathology, Male, Mice, Mice, SCID, Rituximab, Sialic Acid Binding Ig-like Lectin 2, Survival Rate, Transplantation, Heterologous, Antibodies, Monoclonal therapeutic use, Antigens, CD metabolism, Antigens, Differentiation, B-Lymphocyte metabolism, Cell Adhesion Molecules metabolism, Hindlimb, Immunoconjugates therapeutic use, Lectins metabolism, Lymphoma, B-Cell therapy, Paralysis etiology
- Abstract
Purpose: CMC-544 is a CD22-targeted immunoconjugate of calicheamicin and exerts a potent cytotoxic effect against CD22+ B-cell lymphoma. This study evaluated antitumor efficacy of CMC-544 against systemically disseminated B-cell lymphoma., Experimental Design: Scid mice received i.v. injections of CD22+ Ramos B-cell lymphoma cells for their systemic dissemination. CMC-544, G5/44, CD33-targeted CMA-676 (control conjugate) or rituximab were given i.p. 3, 9, 15, or 21 days after B-cell lymphoma dissemination. Diseased mice were monitored daily for hind-limb paralysis and death. Histopathological examination of CMC-544-treated and vehicle-treated diseased mice was also performed., Results: Mice with disseminated B-cell lymphoma developed hind-limb paralysis within 35 days. When given up to 15 days after B-cell lymphoma dissemination, CMC-544 extended survival of the diseased mice to >100 days, and these mice were considered cured. CMC-544 was efficacious when given during both the early initiation phase and the late established phase of the disease. A single dose of CMC-544 was effective in delaying the occurrence of hind-limb paralysis. In contrast, neither CMA-676 nor unconjugated G5/44 was effective. Rituximab was effective when given early in the disease process but not when the disease was established. Histopathological analysis revealed B-cell lymphoma infiltration in brain, spinal cord, bone marrow, and kidney in vehicle-treated but not in CMC-544-treated diseased mice. Consistent with its efficacy against the disseminated B-cell lymphoma, CMC-544 also caused regression of established Ramos B-cell lymphoma xenografts in scid mice., Conclusions: CMC-544 confers strong therapeutic activity against systemic disseminated B-cell lymphoma and protects mice from hind-limb paralysis and death. These results support clinical evaluation of CMC-544 in the treatment of CD22+ lymphoid malignancies.
- Published
- 2004
- Full Text
- View/download PDF
32. Antibody-targeted chemotherapy with the calicheamicin conjugate hu3S193-N-acetyl gamma calicheamicin dimethyl hydrazide targets Lewisy and eliminates Lewisy-positive human carcinoma cells and xenografts.
- Author
-
Boghaert ER, Sridharan L, Armellino DC, Khandke KM, DiJoseph JF, Kunz A, Dougher MM, Jiang F, Kalyandrug LB, Hamann PR, Frost P, and Damle NK
- Subjects
- Animals, Antigens chemistry, Carcinoma metabolism, Cell Line, Tumor, Cell Separation, Collagen chemistry, Dose-Response Relationship, Drug, Dose-Response Relationship, Immunologic, Drug Combinations, Enediynes, Female, Flow Cytometry, Humans, Hydrolysis, Kinetics, Laminin chemistry, Male, Mice, Mice, Nude, Models, Chemical, Neoplasm Transplantation, Protein Binding, Proteoglycans chemistry, Sensitivity and Specificity, Surface Plasmon Resonance, Tissue Distribution, Aminoglycosides chemistry, Aminoglycosides pharmacology, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal pharmacology, Hydrazines pharmacology, Immunotherapy methods, Lewis Blood Group Antigens chemistry
- Abstract
Purpose: Linking a cytotoxic anticancer drug to an antibody that recognizes a tumor-associated antigen can improve the therapeutic index of the drug. We asked whether a conjugate of the cytotoxic antibiotic N-acetyl gamma calicheamicin dimethyl hydrazide (CalichDMH) and an antibody recognizing Lewis(y) (Le(y)) antigen could eliminate carcinomas that express Le(y). Because Le(y) is highly expressed on carcinomas of colon, breast, lung, ovary, and prostate, a CalichDMH conjugate targeting Le(y) could provide a treatment option for various cancers., Experimental Design: The humanized anti-Le(y) antibody hu3S193 was conjugated to CalichDMH via the bifunctional AcBut linker. Selectivity and avidity of the conjugate (hu3S193-CalichDMH) for Le(y)-BSA or Le(y+) cells was tested by BIAcore or flow cytometry. Cytotoxicity of hu3S193-CalichDMH was compared with toxicity of a control conjugate on monolayers of Le(y+) and Le(y-) carcinoma cells. Inhibition of tumor growth by hu3S193-CalichDMH was assessed on three types of s.c. xenografts., Results: Hu3S193-CalichDMH had similar selectivity as hu3S193. The conjugate had lower affinity for Le(y)-BSA but not for Le(y+) cells. When tested on monolayers of human Le(y+) carcinoma cells, hu3S193-CalichDMH was more cytotoxic than a control conjugate. This difference in efficacy was not noted on Le(y-) cells. Efficacy of hu3S193-CalichDMH depended on the expression of Le(y) and on the sensitivity of the cells to CalichDMH. In vivo, hu3S193-CalichDMH inhibited growth of xenografted human gastric (N87), colon (LOVO), and prostate carcinomas (LNCaP). When used against N87 xenografts, hu3S193-CalichDMH arrested tumor growth for at least 100 days., Conclusion: Hu3S193-CalichDMH can specifically eliminate Le(y+) tumors. These results support development of this conjugate for treatment of carcinomas.
- Published
- 2004
- Full Text
- View/download PDF
33. Antibody-targeted chemotherapy with CMC-544: a CD22-targeted immunoconjugate of calicheamicin for the treatment of B-lymphoid malignancies.
- Author
-
DiJoseph JF, Armellino DC, Boghaert ER, Khandke K, Dougher MM, Sridharan L, Kunz A, Hamann PR, Gorovits B, Udata C, Moran JK, Popplewell AG, Stephens S, Frost P, and Damle NK
- Subjects
- Animals, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal, Humanized, Antigens, CD metabolism, Antigens, Differentiation, B-Lymphocyte metabolism, Antineoplastic Agents pharmacology, Cell Line, Tumor, Female, Humans, Immunoglobulin G metabolism, Immunotherapy methods, Inhibitory Concentration 50, Inotuzumab Ozogamicin, Lectins metabolism, Lymphoma, B-Cell metabolism, Lymphoma, Non-Hodgkin metabolism, Mice, Mice, Inbred BALB C, Mice, Nude, Models, Chemical, Neoplasm Transplantation, Protein Binding, Sialic Acid Binding Ig-like Lectin 2, Time Factors, Antibodies, Monoclonal therapeutic use, Antigens, CD biosynthesis, Antigens, Differentiation, B-Lymphocyte biosynthesis, Cell Adhesion Molecules, Immunoconjugates therapeutic use, Lectins biosynthesis, Lymphoma, B-Cell therapy
- Abstract
Antibody-targeted chemotherapy with gemtuzumab ozogamicin (CMA-676, a CD33-targeted immunoconjugate of N-acetyl-gamma-calicheamicin dimethyl hydrazide [CalichDMH], a potent DNA-binding cytotoxic antitumor antibiotic) is a clinically validated therapeutic option for patients with acute myeloid leukemia (AML). Here, we describe the preclinical profile of another immunoconjugate of CalichDMH, CMC-544, targeted to CD22 expressed by B-lymphoid malignancies. CMC-544 comprises a humanized IgG4 anti-CD22 monoclonal antibody (mAb), G5/44, covalently linked to CalichDMH via an acid-labile 4-(4'-acetylphenoxy) butanoic acid (AcBut) linker. Both CMC-544 and unconjugated G5/44 bound human CD22 with subnanomolar affinity. CMC-544, but not unconjugated G5/44, exerted potent cytotoxicity against CD22+ B-cell lymphoma (BCL) cell lines (inhibitory concentration of 50%: 6-600 pM CalichDMH). CMC-544 caused a potent inhibition of growth of small but established BCL xenografts leading to cures (therapeutic index > 10). CMC-544 prevented the establishment of BCL xenografts and also caused regression of large BCLs (> 1.5 g tumor mass). In contrast, unconjugated CalichDMH, unconjugated G5/44, and an isotype-matched control conjugate, CMA-676, were ineffective against these BCL xenografts. Thus, CD22-targeted delivery of CalichDMH is a potent and effective preclinical therapeutic strategy for BCLs. The strong antitumor profile of CMC-544 supports its clinical evaluation as a treatment option for B-lymphoid malignancies.
- Published
- 2004
- Full Text
- View/download PDF
34. Reduced expression of EphrinA1 (EFNA1) inhibits three-dimensional growth of HT29 colon carcinoma cells.
- Author
-
Potla L, Boghaert ER, Armellino D, Frost P, and Damle NK
- Subjects
- Base Sequence, Carcinoma, Cell Division drug effects, Cytoskeletal Proteins metabolism, DNA Primers, Ephrin-A1, Ephrin-A2, Glycosylphosphatidylinositols metabolism, HT29 Cells, Humans, Kinetics, Molecular Sequence Data, Oligodeoxyribonucleotides, Antisense pharmacology, Phosphorylation, Phosphotyrosine metabolism, Polymerase Chain Reaction, Time Factors, Transcription Factors metabolism, Transfection, beta Catenin, Cell Division physiology, Proteins genetics, Trans-Activators
- Abstract
Ephrin A1 (EFNA1) is a GPI-anchored ligand that preferentially binds to the receptor tyrosine kinase, EphA2. EphA2 is over-expressed in malignant melanocytes and in prostate carcinoma cells. Whether activation of EphA2 by EFNA1 is involved in aberrant growth or differentiation of cancer cells is currently not known. We studied the effect of reducing EFNA1 on the growth of a colon carcinoma cell line (HT29). HT29 cells were transfected with EFNA1 antisense yielding clones that expressed less than 25% of EFNA1 found in vector controls. EFNA1-antisense transfectants grew slower than controls when cultured as three-dimensional spheroids. When grown as monolayers, the transfectants had a similar doubling time of the vector controls. These results indicated that autocrine stimulation of EphA2 by EFNA1 could trigger an indirect growth signal by overcoming 'contact inhibition'. Following addition of EFNA1-Fc to HT29 cells, tyrosine hyperphosphorylation of EphA2, E-cadherin, and beta-catenin were observed. Because the function of E-cadherin is associated with contact inhibition of HT29 cells, phosphorylation of E-cadherin and beta-catenin by activation of EphA1 is one possible mechanism by which HT29 cells alleviate contact inhibition.
- Published
- 2002
- Full Text
- View/download PDF
35. A dominant-negative mutant of the platelet-derived growth factor A-chain increases survival of hamsters implanted intracerebrally with the highly invasive CxT24-neo3 glioblastoma cell.
- Author
-
Kaetzel DM, Reid JD 4th, Pedigo N, Zimmer SG, and Boghaert ER
- Subjects
- Animals, Brain Neoplasms genetics, Cricetinae, Glioblastoma genetics, Injections, Intraventricular, Mesocricetus, Neoplasm Invasiveness, Neoplasm Transplantation, Organoids, Platelet-Derived Growth Factor biosynthesis, Platelet-Derived Growth Factor physiology, RNA, Messenger biosynthesis, RNA, Messenger genetics, Transfection, Tumor Cells, Cultured, Brain Neoplasms therapy, Genes, Dominant, Glioblastoma therapy, Platelet-Derived Growth Factor genetics
- Abstract
Evidence is accumulating to suggest a role for PDGF in stimulating malignant growth in astrocytoma, although it has been obtained using model systems (growth in 2-dimensional cell culture, athymic nude mice) that do not assess the complex interactions of these tumors with normal brain tissue. In the current study, the highly invasive hamster glioblastoma cell line CxT24-neo3 was used as a model to study the role of platelet-derived growth factor (PDGF) in mediating malignant growth both in vitro and in vivo when implanted directly into the right lateral ventricle of the brain. Co-expression of PDGF B-chain mRNA and PDGF alpha-receptors was detected in these cells, indicating potential for autocrine activation of their growth. CxT24-neo3 cells transfected with wild-type and receptor binding-deficient forms of the PDGF A- and B-chains displayed alterations in their abilities to grow as three-dimensional spheroids, with overexpression of wild-type B-chain resulting in increased spheroid formation, but a decreased rate of spheroid growth. Influence of these PDGF polypeptides on tumor invasion and survival time in vivo was evaluated following implantation of these spheroids in the brain. While all hamsters implanted with control spheroids died within 21 d (average 17 d), those implanted with cells expressing the receptor binding-deficient A-chain survived for much greater periods of time (average 80 d). Modest increases in survival were also seen in cells stably expressing wild-type A-chain (25 d) and mutant B-chain (26 d) proteins. The present study suggests an important role of PDGF in mediating the malignant growth of the CxT24-neo3 cell line in cerebral cortex, possibly via paracrine interactions with normal cortical cell types (i.e., glia, neurons).
- Published
- 1998
- Full Text
- View/download PDF
36. Distinct patterns of E-cadherin CpG island methylation in papillary, follicular, Hurthle's cell, and poorly differentiated human thyroid carcinoma.
- Author
-
Graff JR, Greenberg VE, Herman JG, Westra WH, Boghaert ER, Ain KB, Saji M, Zeiger MA, Zimmer SG, and Baylin SB
- Subjects
- Carcinoma pathology, DNA Methylation, Genes, Tumor Suppressor genetics, Humans, Thyroid Neoplasms pathology, Tumor Cells, Cultured, Cadherins genetics, Cadherins metabolism, Carcinoma genetics, CpG Islands genetics, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Thyroid Neoplasms genetics
- Abstract
Expression of the invasion/metastasis suppressor, E-cadherin, is diminished or lost in thyroid carcinomas. Yet, mutational inactivation of E-cadherin is rare. Herein, we show that this loss is associated with hypermethylation of the E-cadherin 5' CpG island in a panel of human thyroid cancer cell lines. This aberrant methylation is evident in 83% of papillary thyroid carcinoma, 11% of follicular thyroid carcinoma, 40% of Hurthle's cell carcinoma, and 21% of poorly differentiated thyroid carcinomas. Contrary to previous reports, the majority of these poorly differentiated thyroid carcinomas express E-cadherin, but often within the cytoplasm rather than at the cell surface. Together, our data indicate that the invasion/metastasis suppressor function of E-cadherin is frequently compromised in human papillary, Hurthle's cell, and poorly differentiated thyroid carcinoma by epigenetic and biochemical events.
- Published
- 1998
37. Immunohistochemical analysis of the proapoptotic protein Par-4 in normal rat tissues.
- Author
-
Boghaert ER, Sells SF, Walid AJ, Malone P, Williams NM, Weinstein MH, Strange R, and Rangnekar VM
- Subjects
- Animals, Apoptosis Regulatory Proteins, Carrier Proteins genetics, Endothelium chemistry, Epithelium chemistry, Female, Gene Expression Regulation, Developmental, Male, Mammary Glands, Animal chemistry, Orchiectomy, Organ Specificity, Ovary chemistry, Ovary cytology, Prostate chemistry, RNA, Messenger analysis, Rats, Rats, Sprague-Dawley, Testosterone physiology, Apoptosis physiology, Carrier Proteins analysis, Intracellular Signaling Peptides and Proteins
- Abstract
Prostate apoptosis response 4 (par-4) is a recently identified gene that encodes a transcription factor, Par-4, with a leucine zipper domain. Par-4 protein is constitutively expressed in various cell lines and is functionally required but not sufficient for apoptosis. Induction of Par-4 in cultured cells is found exclusively during apoptosis, and ectopic overexpression of Par-4 enhances the potency of apoptotic stimuli. Western or Northern blot analysis on mRNA or protein extracts, respectively, from rat organs revealed that the expression of Par-4 was ubiquitous and was not restricted to any specific organ(s). To further identify specific cell types that expressed Par-4, we performed an immunohistochemical analysis of the protein in paraffin-embedded sections of various organs from rats. Our findings indicated that consistent with its proapoptotic role, Par-4 is expressed in apoptotic granulosa cells of atretic ovarian follicles and in terminally differentiated cells, such as the cardiomyocytes, cerebellar Purkinje cells, and pyramidal cells of the hypothalamus. Moreover, testosterone ablation by castration of rats caused an early and transient induction of Par-4 in the ductal cells of the prostate that undergo apoptosis. By contrast, in tissues in which the cells could be visually differentiated from their mature counterparts, Par-4 expression was lowest in the mature cells. This was the case for epithelia of the mammary and the prostate gland in which the basal cells maintained higher protein levels of Par-4 than did the terminally differentiated ductal cells. Similarly, cells of the stratum corneum of the skin and cells on top of the duodenal villi stained less intensely for Par-4 as compared to the stem cells in the stratum basale and at the bottom of the crypts of Lieberkühn, respectively. It is possible that Par-4 has to be down-regulated for successful differentiation in these tissues. Taken together, the widespread expression of Par-4 in various adult cell types underscores the physiological importance of the protein. The observation of constitutive Par-4 expression in the stem cell compartments is inconsistent with the probability of apoptosis per se and can be extended to determine whether Par-4 plays a role in other cellular processes.
- Published
- 1997
38. Insulin-like growth factors (IGF) enhance three-dimensional (3D) growth of human glioblastomas.
- Author
-
Morford LA, Boghaert ER, Brooks WH, and Roszman TL
- Subjects
- Humans, Brain Neoplasms pathology, Glioblastoma pathology
- Abstract
Human glioblastomas (gliomas) are characterized as rapidly growing brain tumors which are highly invasive but rarely metastatic. Human gliomas synthesize and secrete increased levels of insulin-like growth factors (IGFs) as well as expressing increased numbers of IGF receptors when compared to normal brain tissue. These observations suggest the existence of an IGF-mediated autocrine mechanism for glioma growth regulation. The purpose of this study was to examine the effect of human recombinant IGF (hrIGF) treatment on the in vitro growth of human glioma monolayer and three-dimensional (3D) multicellular spheroid cultures. The data demonstrate that hrIGF-I treatment of glioma cell lines slightly enhanced tumor monolayer proliferation as measured by [(3)H]thymidine incorporation. In contrast, treatment of glioma spheroids with hrIGF-I or hrDes(1-3)IGF-I, the truncated brain form of IGF-I, dramatically enhanced 3D tumor growth with a 1.5-2-fold reduction in spheroid doubling time (FRSDT). In addition, IGF-treated glioma spheroids were more densely packed than spheroids grown in media alone with no observed necrosis. These data suggest that IGFs will dramatically enhance glioma proliferation when 3D cell-cell contact occurs. This observed enhancement suggests that IGFs both synthesized in the brain and systemically support rapid proliferation of gliomas in vivo.
- Published
- 1997
- Full Text
- View/download PDF
39. The effects of group II phospholipase A2 on ras-induced metastasis.
- Author
-
Davis TW, Boghaert ER, Guthridge CJ, Steiner MR, and Zimmer SG
- Subjects
- Animals, Arachidonic Acid metabolism, Cell Line, Fibroblasts, Genetic Vectors, Neoplasm Metastasis, Neoplasms, Experimental pathology, Phospholipases A2, Rats, Rats, Inbred F344, Recombinant Proteins metabolism, Transfection, Cell Transformation, Neoplastic, Genes, ras, Phospholipases A metabolism
- Published
- 1997
- Full Text
- View/download PDF
40. Quantitative and qualitative differences in growth, invasion and lung colonization of an anaplastic and a papillary human thyroid cancer cell line in vitro and in vivo.
- Author
-
Boghaert ER, Ain K, Taylor K, Greenberg VL, Fowler C, and Zimmer SG
- Subjects
- Animals, Basement Membrane pathology, Cell Division, Gelatinases metabolism, Humans, Lung Neoplasms secondary, Mice, Mice, Nude, Neoplasm Invasiveness, Neoplasm Metastasis, Tumor Cells, Cultured, Carcinoma pathology, Carcinoma, Papillary pathology, Thyroid Neoplasms pathology
- Abstract
Invasion and metastasis remain major reasons for failure of anti-cancer therapy. Cell lines derived from human carcinomas are frequently used to investigate the molecular mechanisms that underlie invasion and metastasis. Unfortunately many of these cell lines do not retain the malignant characteristics of their parental tumors. We therefore conducted a series of experiments in vivo and in vitro to identify which aspects of malignancy of a papillary (NPA'87) and an anaplastic (DR090-1) thyroid carcinoma were consistent with the pathology of the parental tumor types. We evaluated tumor growth, invasion and metastasis of DRO90-1 and NPA'87 in vivo following inoculation of the tumor cells under the dermis, under the renal capsule and into the lateral tail vein of nude mice. This evaluation in vivo showed that the anaplastic carcinoma had a faster growth rate compared with the papillary carcinoma. Furthermore, the papillary carcinoma cells could destroy and infiltrate surrounding tissue but were not capable of extravasation and colonization of lung tissue. The anaplastic cells formed lung nodules following injection into the tail vein of nude mice. This lung colonizing capability of DRO90-1 correlated with their capacity to secrete an active 62 kDa gelatinase and to migrate through reconstituted basement membrane in vitro.
- Published
- 1996
- Full Text
- View/download PDF
41. Reduction of translation initiation factor 4E decreases the malignancy of ras-transformed cloned rat embryo fibroblasts.
- Author
-
Graff JR, Boghaert ER, De Benedetti A, Tudor DL, Zimmer CC, Chan SK, and Zimmer SG
- Subjects
- Animals, Cell Line, Eukaryotic Initiation Factor-4E, Fibroblasts, Mice, NM23 Nucleoside Diphosphate Kinases, Neoplasm Invasiveness, Neoplasm Metastasis, Ornithine Decarboxylase biosynthesis, Protein Biosynthesis, Rats, Transcription Factors biosynthesis, Cell Transformation, Neoplastic, Genes, ras, Monomeric GTP-Binding Proteins, Nucleoside-Diphosphate Kinase, Peptide Initiation Factors physiology
- Abstract
Expression of the T24ras oncogene induces malignancy (tumor growth, invasion and metastasis) in cloned rat embryo fibroblasts (CREF T24). In CREF T24, the rate of phosphorylation of eukaryotic translation initiation factor 4E (eIF-4E) is increased, resulting in increased protein synthesis rates. We have recently shown that reducing the protein levels of eIF-4E in CREF T24 (AS4E line) markedly decreases soft-agar colonization, increases tumor latency periods and increases tumor doubling times without significantly altering monolayer growth. In this study, cells with reduced eIF-4E had delayed and reduced invasiveness and decreased experimental metastasis. Furthermore, reduced eIF-4E levels correlated with decreased expression of the metastasis-associated 92-kDa collagenase type-IV and exon-6 variants of the CD44 adhesion molecule [CD44(6v)]. Reduced eIF-4E levels correlated inversely with increased levels of the putative metastasis-suppressor protein nm23. Cell lines established from AS4E tumors and lung metastases exhibited increased levels of eIF-4E protein and protein synthesis rates compared to the AS4E line. Tumor-derived AS4E had the shortened tumor latency periods of CREF T24 but displayed the slow tumor-growth rates of AS4E. Tumor-derived AS4E exhibited the metastatic capacity of CREF T24 controls. Furthermore, tumor- and lung-nodule-derived AS4E expressed levels of CD44 (6v) and the 92-kDa collagenase type IV comparable to CREF T24 and displayed reduced levels of nm23 relative to AS4E. These results demonstrate that eIF-4E is an important effector molecule involved in oncogenic p21ras-induced malignant transformation.
- Published
- 1995
- Full Text
- View/download PDF
42. Inhibition of collagenolytic activity relates to quantitative reduction of invasion in vitro in a c-Ha-ras transfected glial cell line.
- Author
-
Boghaert ER, Chan SK, Zimmer C, Grobelny D, Galardy RE, Vanaman TC, and Zimmer SG
- Subjects
- Animals, Basement Membrane pathology, Cell Line, Transformed, Cell Movement drug effects, Chick Embryo, Collagen, Cricetinae, Culture Media, Conditioned chemistry, Culture Media, Serum-Free, Drug Combinations, Gelatinases metabolism, Heart embryology, Laminin, Mesocricetus, Myocardium pathology, Neoplasm Proteins metabolism, Neuroglia enzymology, Proteoglycans, Recombinant Fusion Proteins, Transfection, Tyrosine pharmacology, Amides pharmacology, Dipeptides pharmacology, Gelatinases antagonists & inhibitors, Neoplasm Invasiveness, Neoplasm Proteins antagonists & inhibitors, Neuroglia pathology, Protease Inhibitors pharmacology, Proto-Oncogene Proteins p21(ras) physiology, Tyrosine analogs & derivatives
- Abstract
The function of proteases in brain tumor invasion is currently not well established. For tumors of epithelial and fibromatous origin collagenase production can enhance the invasive capacity of cells to penetrate basement membranes. We showed previously that a c-Ha-ras transformed glial cell line (CxT24neo3) invaded hamster brain tissue in vivo. These cells were also capable of invading reconstituted basement membrane and embryonic chick hearts in vitro. Since the histopathology of CxT24neo3 tumors mimics that of glioblastoma multiforme in humans, CxT24neo3 was used as the model in vitro for this type of brain tumor. Presently, we detected by zymogram analysis a gelatinase that was secreted by CxT24neo3 and that had an apparent molecular mass of 62 kD. To verify whether gelatinase affected invasion in vitro of these glial cells we determined the efficacy of a substrate specific collagenase inhibitor on invasion in vitro. GM6001 is a synthetic polypeptide that specifically occupies the substrate binding sites of metalloprotease. Since this drug did not show cytotoxicity, its specificity for metalloprotease is a valuable tool to evaluate the physiological function of these enzymes on invasion. We found that treatment of CxT24neo3 with GM6001 reduced the fraction of invading CxT24neo3 cells through reconstituted basement membrane. These data suggest that metalloproteases can stimulate brain tumor invasion.
- Published
- 1994
- Full Text
- View/download PDF
43. Increasing N-CAM-mediated cell-cell adhesion does not reduce invasion of RSV-transformed WC5 rat cerebellar cells.
- Author
-
Brady-Kalnay SM, Boghaert ER, Zimmer S, and Brackenbury R
- Subjects
- Animals, Avian Sarcoma Viruses, Cell Adhesion Molecules, Neuronal chemistry, Cell Adhesion Molecules, Neuronal genetics, Cell Line, Transformed, Cerebellum metabolism, Collagen, Drug Combinations, Laminin, Molecular Weight, Proteoglycans, Rats, Temperature, Transfection, Cell Adhesion, Cell Adhesion Molecules, Neuronal metabolism, Cerebellum pathology, Neoplasm Invasiveness
- Abstract
The WC5 rat cerebellar cell line, infected with a Rous sarcoma virus (RSV) that is temperature-sensitive for pp60v-src transformation, expresses high levels of the neural cell adhesion molecule, N-CAM, when grown at the non-permissive temperature for pp60v-src activity. At the permissive temperature, N-CAM expression is 4- to 10-fold reduced and the cells aggregate poorly. To evaluate the effects of variations in N-CAM expression, we compared the invasive ability of transformed WC5 cells that express low levels of N-CAM with transformed cells in which N-CAM-mediated adhesion was restored. WC5 cells were transfected with expression vectors containing cDNAs encoding the 120 or 180 kDa forms of chicken N-CAM linked to constitutive promoters. Several permanently transfected lines that expressed chicken N-CAM at the cell surface were isolated. These cell lines showed enhanced aggregation at the permissive temperature relative to untransfected WC5 cells or cells transfected with control constructs. By comparing the ability of control and transfected WC5 cells to invade reconstituted extracellular matrix, we tested the effect of variations in N-CAM-mediated adhesion on invasion. Clones that expressed high levels of N-CAM showed invasion rates that were similar to control cells, indicating that increasing N-CAM-mediated adhesion does not inhibit the invasiveness of RSV-transformed WC5 cells.
- Published
- 1993
- Full Text
- View/download PDF
44. Invasion in vitro of malignant hamster brain tumor cells is influenced by the number of cells and the mode of malignant progression.
- Author
-
Boghaert ER, Simpson JF, and Zimmer SG
- Subjects
- Animals, Cells, Cultured, Cerebral Cortex, Cricetinae, Kinetics, Mathematics, Mesocricetus, Models, Theoretical, Transfection, Brain Neoplasms pathology, Cell Transformation, Neoplastic, Genes, ras, Glioma pathology, Neoplasm Invasiveness pathology, Neoplasm Metastasis pathology
- Abstract
We investigated the capacity of two glial tumor cell lines (CxT24neo3 and CxT3Cl5) to invade through reconstituted basement membrane (Matrigel, MG). The purpose of our experiments was to establish whether the number of cells or the mode of malignant progression would quantitatively modify the invasion of a brain tumor cell population. To accomplish this goal, we used a vital-dye method to assess the fraction of cells that invaded through 30 micrograms MG coated on a polycarbonate filter (8 microns pore size). Our experiments demonstrated that the fraction of invasive CxT24neo3 and CxT3Cl5 cells in vitro reproducibly differed as a function of the number of initially seeded cells. This showed that invasion through MG was subject to quantitative changes caused by the number of cells present. Since CxT24neo3 and CxT3Cl5 became malignant by transfection with different oncogenes, the results also indicated that the type of quantitative change was influenced by the mode of malignant progression.
- Published
- 1992
45. Invasion by WC5 rat cerebellar cells is independent of RSV-induced changes in growth and adhesion.
- Author
-
Brady-Kalnay SM, Boghaert ER, Zimmer S, Soll DR, and Brackenbury R
- Subjects
- Animals, Avian Sarcoma Viruses, Cell Aggregation physiology, Cell Communication physiology, Cell Line, Transformed, Cell Transformation, Viral, Cerebellum pathology, Chick Embryo, Extraembryonic Membranes pathology, Neoplasm Invasiveness pathology, Rats, Temperature, Cell Adhesion physiology, Cell Adhesion Molecules, Neuronal metabolism, Cell Movement physiology, Neoplasm Invasiveness physiopathology
- Abstract
The WC5 rat cerebellar cell line, which is infected with a Rous sarcoma virus that is temperature-sensitive for pp60src transformation, shows temperature-dependent expression of the neural-cell-adhesion molecule (N-CAM) and glial fibrillary acidic protein (GFAP). We found that WC5 cells maintained at the non-permissive temperature in both monolayer cultures and spheroids are subject to density-dependent inhibition of growth, whereas cells maintained at the permissive temperature continued to grow. The movement of isolated WC5 cells at both temperatures was similar, while the migration of WC5 cells out of 3-dimensional aggregates was faster at the non-permissive temperature. We tested whether the RSV-induced changes affect the invasion of the WC5 cells in 2 in vitro assays: the chorio-allantoic-membrane assay and the chick-heart-fragment assay. In both assays, WC5 cells grown at either temperature were invasive. These results indicate that growth rate is unrelated to invasion and that loss of N-CAM-mediated cell-cell adhesion is not necessary for invasion.
- Published
- 1991
- Full Text
- View/download PDF
46. The influence of the presence of adenovirus 5 E1a and E1b sequences on the pathology of rat embryonic fibroblasts transfected with activated c-Ha-ras and v-ras.
- Author
-
Boghaert ER, Austin V, and Zimmer SG
- Subjects
- Animals, Fibroblasts physiology, Humans, Lung Neoplasms genetics, Lung Neoplasms secondary, Lymphatic Metastasis genetics, Neoplasm Invasiveness genetics, Neoplasm Invasiveness pathology, Neoplasm Metastasis genetics, Neoplasm Metastasis pathology, Neoplasm Transplantation, Neoplasms, Experimental genetics, Rats, Rats, Inbred F344, Transfection, Tumor Cells, Cultured, Adenoviruses, Human genetics, Cell Transformation, Viral genetics, Fibroblasts pathology, Genes, ras genetics, Neoplasms, Experimental pathology
- Abstract
We compared the pathology of two groups of tumors following implantation of cells enmeshed in alginate beads into the syngeneic rat. The first group of tumors was generated by implanting alginate beads containing cloned embryonic fibroblasts (CREF) that were transfected with activated c-Ha-ras (T24) and v-ras (pH1) (CREF tumors). The second group was created by implantation of CREF cells that were transfected with E1a and E1b of wild type adenovirus type 5 prior to transfection with T24 and pH1 (Wt tumors). Alginate beads were implanted at three different sites in the rat, i.e. subcutaneous in the flank, subcutaneous in the tail and under the renal capsule. Tumorigenicity, invasiveness and metastatic capacity of the transfectant cell lines were determined. The tumor latency period (TLP), the doubling time of the tumors and the metastatic capacity of the cell lines depended on the site of implantation. Invasion was not influenced by site-dependency. Wt tumors were invasive and generally had longer TLP than the CREF tumors. Wt tumors did not metastasize to the lungs as opposed to CREF tumors. We concluded that the genetic background of Wt cells modulated the effect of ras transfection by stretching the TLP and by limiting the metastatic potential to the draining lymph nodes. Malignancy per se was not repressed since no differences in invasive capacity were noticed.
- Published
- 1991
- Full Text
- View/download PDF
47. The effect of dibutyryl camp (dBcAMP) on morphological differentiation, growth and invasion in vitro of a hamster brain-tumor cell line: a comparative study of dBcAMP effects in 2- and 3-dimensional cultures.
- Author
-
Boghaert ER, Simpson J, Jacob RJ, Lacey T, Walsh JW, and Zimmer SG
- Subjects
- Animals, Brain Neoplasms ultrastructure, Cell Division drug effects, Cricetinae, Tumor Cells, Cultured, Brain Neoplasms pathology, Bucladesine pharmacology, Neoplasm Invasiveness
- Abstract
The use of agents that stimulate cancer cells to differentiate is proposed as a potential approach to the treatment of malignancy. To evaluate the effects of a differentiation inducer on morphology, growth and invasion in vitro of brain-tumor cells, a diffusely invasive hamster glial cell line (CxT3C15) was treated with ImM dibutyryl cyclic adenosine monophosphate (dBcAMP). The efficacy of dBcAMP was tested in monolayer cultures, 3-dimensional static cultures (i.e., spheroids) and confrontation cultures with an embryonic chick heart. CxT3C15 cells exhibited increased numbers of long cellular processes (morphological differentiation) following treatment of monolayer cultures with ImM dBcAMP. One mM dBcAMP also altered the macroscopic and ultrastructural morphology of CxT3C15 grown as spheroids. These alterations were: (i) a fast transition of rough to smooth morphology macroscopically, and (ii) fading of the cell borders concomitant with the disappearance of cell-membrane excrescences, as seen by scanning electron microscopy. Exponential growth of CxT3C15 in monolayers was not changed following treatment with ImM dBcAMP. Treatment of CxT3C15 spheroids with the same dose of dBcAMP caused a reduction of relative volume increase (30-40%). Invasion of CxT3C15 in an embryonic chick heart in vitro was not altered after addition (prior to or at the time of co-culture) of ImM dBcAMP to the co-cultures. These results indicate that invasion of CxT3C15 is not necessarily linked to morphological differentiation or moderated by reduced proliferation.
- Published
- 1991
- Full Text
- View/download PDF
48. Tumour invasion and metastasis: therapeutic implications?
- Author
-
Mareel MM, Bracke ME, and Boghaert ER
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Chick Embryo, Female, Flavonoids therapeutic use, Humans, Laminin physiology, Mice, Microtubules drug effects, Microtubules pathology, Neoplasms drug therapy, Neoplasms etiology, Neoplasms genetics, Neoplasms pathology, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neoplasms, Experimental therapy, Oncogenes, Plasminogen Activators physiology, Rats, Neoplasm Invasiveness, Neoplasm Metastasis pathology, Neoplasm Metastasis prevention & control, Neoplasm Metastasis therapy, Neoplasms therapy
- Abstract
Invasion and metastasis is the hallmark of tumour malignancy. Some aspects of invasion and metastasis with potential implications for tumour therapy are reviewed: the value of laboratory models; the acquisition of invasiveness and metastatic capability during carcinogenesis; the relationship between growth and invasion; clinical and experimental anti-invasive and antimetastatic agents. The value of joint experimental and clinical research is underscored.
- Published
- 1986
- Full Text
- View/download PDF
49. Numerical evaluation of the kidney invasion test.
- Author
-
Boghaert ER, Distelmans W, Van Ginckel R, and Mareel MM
- Subjects
- Animals, Cell Line, Transformed, Collagen, Mice, Mice, Inbred C3H, Neoplasm Invasiveness pathology, Subrenal Capsule Assay
- Abstract
We have implanted MO4 transformed mouse cells attached to a collagen matrix (artificial tumor) under the renal capsule of syngeneic mice. It was our purpose to evaluate whether or not the kidney invasion test (KIT) could be used for the determination in vivo of the invasive capacity of cultured cell populations. Invasion was expressed in terms of the proportion of the depth of the invasive part of the tumor to the total tumor thickness (invasion rate), both measured macroscopically after hemisection of the kidney. Macroscopic findings were confirmed by histology. For MO4 cells the invasion index changed in function of time after implantation. The relationship 'invasion index versus time after implantation' was demonstrated to be constant in 3 independent groups of mice. Therefore, we propose determination of the invasion rate versus time in the KIT as a method to compare the invasive capacity in vivo of different cell lines.
- Published
- 1987
50. Qualitative and quantitative analysis of tumour invasion in vivo and in vitro.
- Author
-
Mareel MM, Van Roy FM, Messiaen LM, Boghaert ER, and Bruyneel EA
- Subjects
- Animals, Chick Embryo, Humans, Image Interpretation, Computer-Assisted, Mice, Neoplasm Staging, Neoplasm Invasiveness
- Abstract
Qualitative and quantitative methods for the analysis of invasion in 'natural' and in experimental tumours in vivo and in vitro are reviewed. In human tumours the functional consequences of invasion were evaluated histologically through staging on the basis of depths of invasion and through the presence of tumour cells inside vessels. Antibodies against components of the basement membrane have facilitated the definition of minimal invasion. With new probes derived from oncogene research the search for molecular differences between invasive and non-invasive parts of the tumour has begun. Since the same methods as those used for analysis of natural tumours also apply to experimental tumours in vivo, the major advantage of the latter is the possibility of manipulation. We have described a new mesenterium assay that may permit the selection of invasive cells from non-invasive ones in transfection experiments. Invasion relative to growth as a function of time was quantified in the kidney invasion test. In three-dimensional confrontations between embryonic chick heart fragments and invasive cells, we have used both a subjective grading and a qualitative computer-assisted image analysis of serial histological sections to score invasion. In two-dimensional confrontations supplementary methods could be applied, since such confrontations permitted direct observations on living cultures. In a variety of natural and experimental tumours, ultrastructural analysis, transmigration in two-compartment chambers, and release of metabolic label have demonstrated the role of motility and of lytic activity in tumour invasion.
- Published
- 1987
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.