69 results on '"Gullbo J"'
Search Results
2. Melflufen for relapsed and refractory multiple myeloma
- Author
-
Oriol, Albert, Larocca, A., Leleu, X., Hajek, R., Hassoun, H., Rodríguez-Otero, P., Paner, A., Schjesvold, F.H., Gullbo, J., Richardson, P.G., and Universitat Autònoma de Barcelona
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Phenylalanine ,Relapsed/refractory multiple myeloma ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,Recurrence ,relapsed/refractory multiple myeloma ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Melflufen ,melphalan flufenamide ,medicine ,Overall survival ,Humans ,Pharmacology (medical) ,Antineoplastic Agents, Alkylating ,Melphalan ,Multiple myeloma ,Pharmacology ,business.industry ,Refractory Multiple Myeloma ,General Medicine ,medicine.disease ,Melphalan-flufenamide ,Survival Rate ,030104 developmental biology ,Novel agents ,030220 oncology & carcinogenesis ,business ,Multiple Myeloma ,Melphalan flufenamide - Abstract
Introduction: The overall survival of patients with multiple myeloma has improved with the advent of novel agents; however, multiple myeloma remains incurable. Combinations of standard-of-care agents such as immunomodulators, proteasome inhibitors, and anti-CD38 monoclonal antibodies are increasingly used in earlier lines of therapy. Patients with disease that is refractory to multiple novel agents represent a population with high unmet medical need and for whom therapies with new mechanisms of action could be beneficial. Melphalan flufenamide (melflufen) has demonstrated encouraging activity in patients with relapsed and refractory multiple myeloma. Areas covered: This review provides an overview of the mechanism of action of melflufen, a first-in-class peptide-drug conjugate that targets aminopeptidases and rapidly delivers alkylating agents into tumor cells. It reviews key Phase I and II clinical trial data for melflufen in combination with dexamethasone as well as in triplet combinations with daratumumab or bortezomib. The safety profile of melflufen, which is characterized primarily by clinically manageable hematologic adverse events, is described. Expert opinion: Melflufen has potential to fill a gap in the myeloma treatment landscape by providing a new mechanism of action with clinically meaningful efficacy and a favorable safety profile in patients refractory to multiple novel agents.
- Published
- 2020
3. Key role of glutamic acid for the cytotoxic activity of the cyclotide cycloviolacin O2
- Author
-
Herrmann, A., Svangård, E., Claeson, P., Gullbo, J., Bohlin, L., and Göransson, U.
- Published
- 2006
- Full Text
- View/download PDF
4. Melflufen: A Peptide-Drug Conjugate for the Treatment of Multiple Myeloma
- Author
-
Mateos, M.V., Bladé, J. (Joan), Bringhen, S. (Sara), Ocio, E.M. (E.), Efebera, Y, Pour, L., Gay, F. (Francesca), Sonneveld, P. (Pieter), Gullbo, J., Richardson, P.G. (Paul Gerard), Mateos, M.V., Bladé, J. (Joan), Bringhen, S. (Sara), Ocio, E.M. (E.), Efebera, Y, Pour, L., Gay, F. (Francesca), Sonneveld, P. (Pieter), Gullbo, J., and Richardson, P.G. (Paul Gerard)
- Abstract
Despite the availability of new therapies that have led to improved outcomes for patients with multiple myeloma, most patients will eventually relapse. With triplet and even quadruplet combination therapies becoming standard in the first and second line, many patients will have few treatment options after second-line treatment. Melflufen (melphalan flufenamide) is a first-in-class peptide–drug conjugate (PDC) that targets aminopeptidases and rapidly releases alkylating agents into tumor cells. Once inside the tumor cells, melflufen is hydrolyzed by peptidases to release alkylator molecules, which become entrapped. Melflufen showed anti-myeloma activity in myeloma cells that were resistant to bortezomib and the alkylator melphalan. In early phase studies (O-12-M1 and HORIZON [OP-106]), melflufen plus dexamethasone has demonstrated encouraging clinical activity and a manageable safety profile in heavily pretreated patients with relapsed/refractory multiple myeloma, including those with triple-class refractory disease and extramedullary disease. The Phase III OCEAN study (OP-104) is further evaluating melflufen plus dexamethasone in patients with relapsed/refractory multiple myeloma. The safety profile of melflufen is characterized primarily by clinically manageable hematologic adverse events. Melflufen, with its novel mechanism of action, has the potential to provide clinically meaningful benefits to patients with relapsed/refractory multiple myeloma, including those with high unmet needs.
- Published
- 2020
- Full Text
- View/download PDF
5. Melflufen: A Peptide-Drug Conjugate for the Treatment of Multiple Myeloma
- Author
-
Mateos, MV, Blade, J, Bringhen, S, Ocio, EM, Efebera, Y, Pour, L, Gay, F, Sonneveld, Pieter, Gullbo, J, Richardson, PG, Mateos, MV, Blade, J, Bringhen, S, Ocio, EM, Efebera, Y, Pour, L, Gay, F, Sonneveld, Pieter, Gullbo, J, and Richardson, PG
- Published
- 2020
6. Small, novel proteins from the mistletoe Phoradendron tomentosum exhibit highly selective cytotoxicity to human breast cancer cells
- Author
-
Johansson, S., Gullbo, J., Lindholm, P., Ek, B., Thunberg, E., Samuelsson, G., Larsson, R., Bohlin, L., and Claeson, P.
- Published
- 2003
- Full Text
- View/download PDF
7. A randomized, multicenter, open-label controlled phase 2 trial of Foxy-5 as neoadjuvant therapy in patients with WNT5A negative colon cancer
- Author
-
Vermorken, J., primary, Cervantes, A., additional, Morsing, P., additional, Johansson, K., additional, Andersson, T., additional, Roest, N. Lindland, additional, Gullbo, J., additional, and Salazar, R., additional
- Published
- 2019
- Full Text
- View/download PDF
8. Abstract P1-12-09: Clinical characteristics of CNS metastases of different breast cancer subtypes – Results from a cohort study
- Author
-
Lindman, H, primary, Nilsson, A, additional, and Gullbo, J, additional
- Published
- 2017
- Full Text
- View/download PDF
9. APR-246 overcomes resistance to cisplatin and doxorubicin in ovarian cancer cells
- Author
-
Mohell, N, primary, Alfredsson, J, additional, Fransson, Å, additional, Uustalu, M, additional, Byström, S, additional, Gullbo, J, additional, Hallberg, A, additional, Bykov, V J N, additional, Björklund, U, additional, and Wiman, K G, additional
- Published
- 2015
- Full Text
- View/download PDF
10. Drug screen in patient cells suggests quinacrine to be repositioned for treatment of acute myeloid leukemia
- Author
-
Eriksson, A, primary, Österroos, A, additional, Hassan, S, additional, Gullbo, J, additional, Rickardson, L, additional, Jarvius, M, additional, Nygren, P, additional, Fryknäs, M, additional, Höglund, M, additional, and Larsson, R, additional
- Published
- 2015
- Full Text
- View/download PDF
11. SHP1 EXPRESSION IS EPIGENETICALLY REGULATED AND INFLUENCES THE SENSITIVITY TO CHEMOTHERAPEUTIC AGENTS IN GLIOBLASTOMA CELLS
- Author
-
Sooman, L., Ekman, S., Bergqvist, M., Gullbo, J., Bergstrom, S., Johansson, Mikael, Wu, X., Blomquist, E., Lennartsson, J., Sooman, L., Ekman, S., Bergqvist, M., Gullbo, J., Bergstrom, S., Johansson, Mikael, Wu, X., Blomquist, E., and Lennartsson, J.
- Published
- 2012
12. Hsp90 is expressed and represents a therapeutic target in human oesophageal cancer using the inhibitor 17-allylamino-17-demethoxygeldanamycin
- Author
-
Wu, X., Wanders, A., Wardega, P., Tinge, B., Gedda, L., Bergstrom, S., Sooman, L., Gullbo, J., Bergqvist, M., Hesselius, P., Lennartsson, J., Ekman, S., Wu, X., Wanders, A., Wardega, P., Tinge, B., Gedda, L., Bergstrom, S., Sooman, L., Gullbo, J., Bergqvist, M., Hesselius, P., Lennartsson, J., and Ekman, S.
- Abstract
Heat shock protein 90 (Hsp90) has been demonstrated to protect oncogenic variants of signalling molecules from degradation and may consequently serve as a therapeutic target for the treatment of oesophageal cancer for which adequate therapy is often lacking. We studied the expression of Hsp90 in tumour tissues of human oesophageal cancer and the impact of Hsp90 inhibition on oesophageal cancer cell lines using the drug 17-allylamino-17-demethoxygeldanamycin (17-AAG). Quantitative immunohistochemistry was performed on formalin-fixed paraffin-embedded tissues from patients with oesophageal cancer. In squamous cell carcinoma, a marked upregulation of Hsp90 could be noted in dysplastic epithelium and invasive cancer compared with normal epithelium. In adenocarcinoma, Hsp90 was expressed in neoplastic epithelium and also in normal non-neoplastic glands weakly. The inhibition of Hsp90 using 17-AAG led to a significant decrease in cell proliferation and viability in human oesophageal cancer cell lines. Using a clonogenic cell survival assay, Hsp90 inhibition significantly sensitised the cells for gamma-photon irradiation. Heat shock protein 90 was found to be critical for proper signalling induced by both epidermal growth factor and insulin-like growth factor-1, in which the inhibition of signalling by 17-AAG correlated with the observed reduction in cell proliferation and viability. These results showed that Hsp90 was selectively expressed in oesophageal cancer tissue compared with the corresponding normal tissue, and the inhibition of Hsp90 resulted in decreased proliferation and viability as well as radiosensitisation of oesophageal cancer cells. Heat shock protein 90 represents a potential therapeutic target in the treatment of patients with oesophageal cancer, alone or in combination with radiotherapy., Wu, X Wanders, A Wardega, P Tinge, B Gedda, L Bergstrom, S Sooman, L Gullbo, J Bergqvist, M Hesselius, P Lennartsson, J Ekman, S eng Research Support, Non-U.S. Gov't England 2009/01/15 09:00 Br J Cancer. 2009 Jan 27;100(2):334-43. doi: 10.1038/sj.bjc.6604855. Epub 2009 Jan 13.
- Published
- 2009
13. The novel melphalan prodrug J1 inhibits neuroblastoma growth in vitro and in vivo
- Author
-
Wickstrom, M., Johnsen, J.I., Ponthan, F., Segerstrom, L., Sveinbjornsson, B., Lindskog, M., Lovborg, Henrik, Viktorsson, K., Lewensohn, R., Kogner, P., Larsson, R., Gullbo, J., Wickstrom, M., Johnsen, J.I., Ponthan, F., Segerstrom, L., Sveinbjornsson, B., Lindskog, M., Lovborg, Henrik, Viktorsson, K., Lewensohn, R., Kogner, P., Larsson, R., and Gullbo, J.
- Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood. The activity of J1 (L-melphalanyl-p-L-fluorophenylalanine ethyl ester), an enzymatically activated melphalan prodrug, was evaluated in neuroblastoma models in vitro and in vivo. Seven neuroblastoma cell lines with various levels of drug resistance were screened for cytotoxicity of J1 alone or in combination with standard cytotoxic drugs, using a fluorometric cytotoxicity assay. J1 displayed high cytotoxic activity in vitro against all neuroblastoma cell lines, with IC50 values in the submicromolar range, significantly more potent than melphalan. The cytotoxicity of J1, but not melphalan, could be significantly inhibited by the aminopeptidase inhibitor bestatin. J1 induced caspase-3 cleavage and apoptotic morphology, had additive effects in combination with doxorubicin, cyclophosphamide, carboplatin, and vincristine, and synergistically killed otherwise drug-resistant cells when combined with etoposide. Athymic rats and mice carrying neuroblastoma xenografts [SHSY5Y, SK-N-BE(2)] were treated with equimolar doses of melphalan, J1, or no drug, and effects on tumor growth and tissue morphology were analyzed. Tumor growth in vivo was significantly inhibited by J1 compared with untreated controls. Compared with melphalan, J1 more effectively inhibited the growth of mice with SH-SY5Y xenografts, was associated with higher caspase-3 activation, fewer proliferating tumor cells, and significantly decreased mean vascular density. In conclusion, the melphalan prodrug J1 is highly active in models of neuroblastoma in vitro and in vivo, encouraging further clinical development in this patient group. Copyright © 2007 American Association for Cancer Research.
- Published
- 2007
- Full Text
- View/download PDF
14. Cytotoxic cyclotides from Viola tricolor
- Author
-
Svangård, E., Göransson, U., Hocaoglu, Z., Gullbo, J., Larsson, R., Claeson, P., Bohlin, L., Svangård, E., Göransson, U., Hocaoglu, Z., Gullbo, J., Larsson, R., Claeson, P., and Bohlin, L.
- Published
- 2004
15. Hsp90 is expressed and represents a therapeutic target in human oesophageal cancer using the inhibitor 17-allylamino-17-demethoxygeldanamycin
- Author
-
Wu, X, primary, Wanders, A, additional, Wardega, P, additional, Tinge, B, additional, Gedda, L, additional, Bergstrom, S, additional, Sooman, L, additional, Gullbo, J, additional, Bergqvist, M, additional, Hesselius, P, additional, Lennartsson, J, additional, and Ekman, S, additional
- Published
- 2009
- Full Text
- View/download PDF
16. Identification of molecular mechanisms for cellular drug resistance by combining drug activity and gene expression profiles
- Author
-
Rickardson, L, primary, Fryknäs, M, additional, Dhar, S, additional, Lövborg, H, additional, Gullbo, J, additional, Rydåker, M, additional, Nygren, P, additional, Gustafsson, M G, additional, Larsson, R, additional, and Isaksson, A, additional
- Published
- 2005
- Full Text
- View/download PDF
17. A nonclonogenic cytotoxicity assay using primary cultures of patient tumor cells for anticancer drug screening
- Author
-
Dhar, S, Gullbo, J, Nilsson, K, Nygren, P, Larsson, R, Dhar, S, Gullbo, J, Nilsson, K, Nygren, P, and Larsson, R
- Published
- 1998
18. Key role of glutamic acid for the cytotoxic activity of the cyclotide cycloviolacin O2
- Author
-
Herrmann, A., Svangård, E., Claeson, P., Gullbo, J., Bohlin, L., Göransson, U., Herrmann, A., Svangård, E., Claeson, P., Gullbo, J., Bohlin, L., and Göransson, U.
19. Anti-tumour activity of the cyclotide cycloviolacin O2 on human cancer cell lines in a hollow fibre model
- Author
-
Svangård, E., Hassan, S., Göransson, U., Herrmann, A., Clark, R., Craik, D., Bohlin, L., Claeson, P., Gullbo, J., Svangård, E., Hassan, S., Göransson, U., Herrmann, A., Clark, R., Craik, D., Bohlin, L., Claeson, P., and Gullbo, J.
20. Structure-activity relationship analysis of cytotoxic cyanoguanidines: selection of CHS 828 as candidate drug
- Author
-
Gullbo Joachim, Burman Robert, and Lövborg Henrik
- Subjects
Medicine ,Biology (General) ,QH301-705.5 ,Science (General) ,Q1-390 - Abstract
Abstract Background N-(6-(4-chlorophenoxy)hexyl)-N'-cyano-N''-4-pyridyl guanidine) (CHS 828) is the first candidate drug from a novel group of anti-tumour agents – the pyridyl cyanoguanidines, shown to be potent compounds interfering with cellular metabolism (inhibition of nicotinamide phosphoribosyl transferase) and NF-κB signalling. Substituted cyanoguanidines are also found in anti-hypertensive agents such as the potassium channel opener pinacidil (N-cyano-N'-(4-pyridyl)-N''-(1,2,2-trimethylpropyl)guanidine) and histamine-II receptor antagonists (e.g. cimetidine, N-cyano-N'-methyl-N''-[2-[[(5-methylimidazol-4-yl]methyl]thio]ethyl)guanidine). In animal studies, CHS 828 has shown very promising activity, and phase I and II studies resulted in further development of a with a water soluble prodrug. Findings To study the structural requirements for cyanoguanidine cytotoxicity a set of 19 analogues were synthesized. The cytotoxic effects were then studied in ten cell lines selected for different origins and mechanisms of resistance, using the fluorometric microculture cytotoxicity assay (FMCA). The compounds showed varying cytotoxic activity even though the dose-response curves for some analogues were very shallow. Pinacidil and cimetidine were found to be non-toxic in all ten cell lines. Starting with cyanoguanidine as the crucial core it was shown that 4-pyridyl substitution was more efficient than was 3-pyridyl substitution. The 4-pyridyl cyanoguanidine moiety should be linked by an alkyl chain, optimally a hexyl, heptyl or octyl chain, to a bulky end group. The exact composition of this end group did not seem to be of crucial importance; when the end group was a mono-substituted phenyl ring it was shown that the preferred position was 4-substitution, followed by 3- and, finally, 2-substitution as the least active. Whether the substituent was a chloro, nitro or methoxy substituent seemed to be of minor importance. Finally, the activity patterns in the ten cell lines were compared. Substances with similar structures correlated well, whilst substances with large differences in molecular structure demonstrated lower correlation coefficients. Conclusion According to this structure-activity relationship (SAR) study, CHS 828 meets the requirements for optimal cytotoxic activity for this class of compounds.
- Published
- 2009
- Full Text
- View/download PDF
21. P-133 - A randomized, multicenter, open-label controlled phase 2 trial of Foxy-5 as neoadjuvant therapy in patients with WNT5A negative colon cancer.
- Author
-
Vermorken, J., Cervantes, A., Morsing, P., Johansson, K., Andersson, T., Roest, N. Lindland, Gullbo, J., and Salazar, R.
- Subjects
- *
NEOADJUVANT chemotherapy , *COLON cancer - Published
- 2019
- Full Text
- View/download PDF
22. Melphalan flufenamide inhibits osteoclastogenesis by suppressing proliferation of monocytes.
- Author
-
Byrgazov K, Lind T, Rasmusson AJ, Andersson C, Slipicevic A, Lehmann F, Gullbo J, Melhus H, Larsson R, and Fryknäs M
- Abstract
Myeloma bone disease is a major complication in multiple myeloma affecting quality of life and survival. It is characterized by increased activity of osteoclasts, bone resorbing cells. Myeloma microenvironment promotes excessive osteoclastogenesis, a process of production of osteoclasts from their precursors, monocytes. The effects of two anti-myeloma drugs, melphalan flufenamide (melflufen) and melphalan, on the activity and proliferation of osteoclasts and their progenitors, monocytes, were assessed in this study. In line with previous research, differentiation of monocytes was associated with increased expression of genes encoding DNA damage repair proteins. Hence monocytes were more sensitive to DNA damage-causing alkylating agents than their differentiated progeny, osteoclasts. In addition, differentiated progeny of monocytes showed increased gene expression of immune checkpoint ligands which may potentially create an immunosuppressive microenvironment. Melflufen was ten-fold more active than melphalan in inhibiting proliferation of osteoclast progenitors. Furthermore, melflufen was also superior to melphalan in inhibition of osteoclastogenesis and bone resorption. These results demonstrate that melflufen may exert beneficial effects in patients with multiple myeloma such as reducing bone resorption and immunosuppressive milieu by inhibiting osteoclastogenesis., Competing Interests: KB, AS, FL are employed by Oncopeptides AB. FL, JG, RL have an equity in Oncopeptides AB. JG and RL are founders of Oncopeptides AB. JG is a consultant for Oncopeptides AB. JG is founder of Theradex Oncology. All other authors declare no conflict of interests., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
23. Melflufen: A Peptide-Drug Conjugate for the Treatment of Multiple Myeloma.
- Author
-
Mateos MV, Bladé J, Bringhen S, Ocio EM, Efebera Y, Pour L, Gay F, Sonneveld P, Gullbo J, and Richardson PG
- Abstract
Despite the availability of new therapies that have led to improved outcomes for patients with multiple myeloma, most patients will eventually relapse. With triplet and even quadruplet combination therapies becoming standard in the first and second line, many patients will have few treatment options after second-line treatment. Melflufen (melphalan flufenamide) is a first-in-class peptide-drug conjugate (PDC) that targets aminopeptidases and rapidly releases alkylating agents into tumor cells. Once inside the tumor cells, melflufen is hydrolyzed by peptidases to release alkylator molecules, which become entrapped. Melflufen showed anti-myeloma activity in myeloma cells that were resistant to bortezomib and the alkylator melphalan. In early phase studies (O-12-M1 and HORIZON [OP-106]), melflufen plus dexamethasone has demonstrated encouraging clinical activity and a manageable safety profile in heavily pretreated patients with relapsed/refractory multiple myeloma, including those with triple-class refractory disease and extramedullary disease. The Phase III OCEAN study (OP-104) is further evaluating melflufen plus dexamethasone in patients with relapsed/refractory multiple myeloma. The safety profile of melflufen is characterized primarily by clinically manageable hematologic adverse events. Melflufen, with its novel mechanism of action, has the potential to provide clinically meaningful benefits to patients with relapsed/refractory multiple myeloma, including those with high unmet needs.
- Published
- 2020
- Full Text
- View/download PDF
24. Targeting aggressive osteosarcoma with a peptidase-enhanced cytotoxic melphalan flufenamide.
- Author
-
Byrgazov K, Anderson C, Salzer B, Bozsaky E, Larsson R, Gullbo J, Lehner M, Lehmann F, Slipicevic A, Kager L, Fryknäs M, and Taschner-Mandl S
- Abstract
Background: Low survival rates in metastatic high-grade osteosarcoma (HGOS) have remained stagnant for the last three decades. This study aims to investigate the role of aminopeptidase N (ANPEP) in HGOS progression and its targeting with a novel lipophilic peptidase-enhanced cytotoxic compound melphalan flufenamide (melflufen) in HGOS., Methods: Meta-analysis of publicly available gene expression datasets was performed to determine the impact of ANPEP gene expression on metastasis-free survival of HGOS patients. The efficacy of standard-of-care anti-neoplastic drugs and a lipophilic peptidase-enhanced cytotoxic conjugate melflufen was investigated in patient-derived HGOS ex vivo models and cell lines. The kinetics of apoptosis and necrosis induced by melflufen and doxorubicin were compared. Anti-neoplastic effects of melflufen were investigated in vivo ., Results: Elevated ANPEP expression in diagnostic biopsies of HGOS patients was found to significantly reduce metastasis-free survival. In drug sensitivity assays, melflufen has shown an anti-proliferative effect in HGOS ex vivo samples and cell lines, including those resistant to methotrexate, etoposide, doxorubicin, and PARP inhibitors. Further, HGOS cells treated with melflufen displayed a rapid induction of apoptosis and this sensitivity correlated with high expression of ANPEP . In combination treatments, melflufen demonstrated synergy with doxorubicin in killing HGOS cells. Finally, Melflufen displayed anti-tumor growth and anti-metastatic effects in vivo ., Conclusion: This study may pave the way for use of melflufen as an adjuvant to doxorubicin in improving the therapeutic efficacy for the treatment of metastatic HGOS., Competing Interests: Conflict of interest statement: KB, AS, FL are employed by Oncopeptides AB; FL and JG have equity in Oncopeptides AB; JG is a founder of Oncopeptides AB and currently provides consultancy to Oncopeptides AB., (© The Author(s), 2020.)
- Published
- 2020
- Full Text
- View/download PDF
25. Analysis of determinants for in vitro resistance to the small molecule deubiquitinase inhibitor b-AP15.
- Author
-
Mofers A, Perego P, Selvaraju K, Gatti L, Gullbo J, Linder S, and D'Arcy P
- Subjects
- Antineoplastic Agents pharmacology, Bortezomib pharmacology, Colonic Neoplasms drug therapy, Glutathione metabolism, Humans, Tumor Cells, Cultured, Apoptosis drug effects, Azepines pharmacology, Benzylidene Compounds pharmacology, Cell Proliferation drug effects, Colonic Neoplasms pathology, Drug Resistance, Neoplasm, Proteasome Inhibitors pharmacology
- Abstract
Background: b-AP15/VLX1570 are small molecule inhibitors of the ubiquitin specific peptidase 14 (USP14) and ubiquitin carboxyl-terminal hydrolase 5 (UCHL5) deubiquitinases (DUBs) of the 19S proteasome. b-AP15/VLX1570 have been shown to be cytotoxic to cells resistant to bortezomib, raising the possibility that this class of drugs can be used as a second-line therapy for treatment-resistant multiple myeloma. Limited information is available with regard to potential resistance mechanisms to b-AP15/VLX1570., Results: We found that b-AP15-induced cell death is cell-cycle dependent and that non-cycling tumor cells may evade b-AP15-induced cell death. Such non-cycling cells may re-enter the proliferative state to form colonies of drug-sensitive cells. Long-term selection of cells with b-AP15 resulted in limited drug resistance (~2-fold) that could be reversed by buthionine sulphoximine, implying altered glutathione (GSH) metabolism as a resistance mechanism. In contrast, drug uptake and overexpression of drug efflux transporters were found not to be associated with b-AP15 resistance., Conclusions: The proteasome DUB inhibitors b-AP15/VLX1570 are cell cycle-active. The slow and incomplete development of resistance towards these compounds is an attractive feature in view of future clinical use., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
26. A novel tumor spheroid model identifies selective enhancement of radiation by an inhibitor of oxidative phosphorylation.
- Author
-
Karlsson H, Senkowski W, Fryknäs M, Mansoori S, Linder S, Gullbo J, Larsson R, and Nygren P
- Abstract
There is a need for preclinical models that can enable identification of novel radiosensitizing drugs in clinically relevant high-throughput experiments. We used a new high-throughput compatible total cell kill spheroid assay to study the interaction between drugs and radiation in order to identify compounds with radiosensitizing activity. Experimental drugs were compared to known radiosensitizers and cytotoxic drugs clinically used in combination with radiotherapy. VLX600, a novel iron-chelating inhibitor of oxidative phosphorylation, potentiated the effect of radiation in tumor spheroids in a synergistic manner. This effect was specific to spheroids and not observed in monolayer cell cultures. In conclusion, the total cell kill spheroid assay is a feasible high-throughput method in the search for novel radiosensitizers. VLX600 shows encouraging characteristics for development as a novel radiosensitizer., Competing Interests: CONFLICTS OF INTEREST MF, SL, JG, RL and PN are minor shareholders of Vivolux AB.
- Published
- 2019
- Full Text
- View/download PDF
27. Synergistic effects of combining proteasome inhibitors with chemotherapeutic drugs in lung cancer cells.
- Author
-
Sooman L, Gullbo J, Bergqvist M, Bergström S, Lennartsson J, and Ekman S
- Subjects
- Bortezomib pharmacology, Cell Line, Tumor, Cisplatin pharmacology, Deoxycytidine analogs & derivatives, Deoxycytidine pharmacology, Drug Therapy, Combination, Gefitinib, Humans, Piperidones pharmacology, Quinazolines pharmacology, Vinblastine analogs & derivatives, Vinblastine pharmacology, Vinorelbine, Gemcitabine, Adenocarcinoma drug therapy, Antineoplastic Agents pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Squamous Cell drug therapy, Proteasome Inhibitors pharmacology, Small Cell Lung Carcinoma drug therapy
- Abstract
Background: The prognosis for patients with disseminated lung cancer is poor and current treatments have limited survival benefit as resistance often occurs, and is often associated with significant toxicity. A possible strategy to improve treatment and evade chemoresistance may be to find new combinations of drugs. The aim of this study was to analyze the potential of combining proteasome inhibitors (PIs) with chemotherapeutic drugs used in the routine treatment for lung cancer patients., Results: The median-effect method was applied to the Fluorometric Microculture Cytotoxicity Assay (FMCA) to evaluate effects of combining two different PIs (bortezomib and b-AP15) with clinically used chemotherapeutic drugs representing different mechanisms of action (cisplatin, gefitinib, gemcitabine and vinorelbine) in two lung cancer cell lines (one sensitive and one resistant). Proteasome inhibition in combination with cisplatin, gemcitabine or vinorelbine had synergistic effects in at least one of the tested cell lines. Furthermore, the effect of gefitinib appeared strongly potentiated by the PI in the least resistant lung cancer cell line, although the level of synergy could not be determined with the median-effect method., Conclusions: Combining PIs with cisplatin, gefitinib, gemcitabine or vinorelbine show potential as new combination chemotherapy for the treatment of lung cancer.
- Published
- 2017
- Full Text
- View/download PDF
28. Melflufen - a peptidase-potentiated alkylating agent in clinical trials.
- Author
-
Wickström M, Nygren P, Larsson R, Harmenberg J, Lindberg J, Sjöberg P, Jerling M, Lehmann F, Richardson P, Anderson K, Chauhan D, and Gullbo J
- Abstract
Aminopeptidases like aminopeptidase N (APN, also known as CD13) play an important role not only in normal cellular functioning but also in the development of cancer, including processes like tumor cell invasion, differentiation, proliferation, apoptosis, motility, and angiogenesis. An increased expression of APN has been described in several types of human malignancies, especially those characterized by fast-growing and aggressive phenotypes, suggesting APN as a potential therapeutic target. Melphalan flufenamide ethyl ester (melflufen, previously denoted J1) is a peptidase-potentiated alkylating agent. Melflufen readily penetrates membranes and an equilibrium is rapidly achieved, followed by enzymatic cleavage in aminopeptidase positive cells, which results in trapping of less lipophilic metabolites. This targeting effect results in very high intracellular concentrations of its metabolite melphalan and subsequent apoptotic cell death. This results in a potency increase (melflufen vs melphalan) ranging from 10- to several 100-fold in different in vitro models. Melflufen triggers a rapid, robust, and an irreversible DNA damage which may account for its ability to overcome melphalan-resistance in multiple myeloma cells. Furthermore, anti-angiogenic properties of melflufen have been described. Consequently, it is hypothesized that melflufen could provide better efficacy but no more toxicity than what is achieved with melphalan, an assumption so far supported by experiences from hollow fiber and xenograft studies in rodents as well as by clinical data from patients with solid tumors and multiple myeloma. This review summarizes the current preclinical and clinical knowledge of melflufen., Competing Interests: CONFLICTS OF INTEREST JG, RL and PN are co-founders and minor shareholders of OncoPeptides AB. DC has received research support from OncoPeptides AB. JH, PS, MJ, PR, JG and FL are consultants of OncoPeptides AB, and JL is CEO of the company. JH, FL, JG, PN, RL and JL have stock options in the company. JL and JH are shareholders.
- Published
- 2017
- Full Text
- View/download PDF
29. Mechanistic characterization of a copper containing thiosemicarbazone with potent antitumor activity.
- Author
-
Karlsson H, Fryknäs M, Strese S, Gullbo J, Westman G, Bremberg U, Sjöblom T, Pandzic T, Larsson R, and Nygren P
- Subjects
- Animals, Antineoplastic Agents chemistry, Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, Dose-Response Relationship, Drug, Gene Expression Regulation, Neoplastic drug effects, Humans, Mice, Oxidative Stress drug effects, Proteasome Endopeptidase Complex metabolism, Pyridines chemistry, Thiosemicarbazones chemistry, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Copper chemistry, Pyridines pharmacology, Thiosemicarbazones pharmacology
- Abstract
Background: The thiosemicarbazone CD 02750 (VLX50) was recently reported as a hit compound in a phenotype-based drug screen in primary cultures of patient tumor cells. We synthesized a copper complex of VLX50, denoted VLX60, and characterized its antitumor and mechanistic properties., Materials and Methods: The cytotoxic effects and mechanistic properties of VLX60 were investigated in monolayer cultures of multiple human cell lines, in tumor cells from patients, in a 3-D spheroid cell culture system and in vivo and were compared with those of VLX50., Results: VLX60 showed ≥ 3-fold higher cytotoxic activity than VLX50 in 2-D cultures and, in contrast to VLX50, retained its activity in the presence of additional iron. VLX60 was effective against non-proliferative spheroids and against tumor xenografts in vivo in a murine model. In contrast to VLX50, gene expression analysis demonstrated that genes associated with oxidative stress were considerably enriched in cells exposed to VLX60 as was induction of reactive oxygen. VLX60 compromised the ubiquitin-proteasome system and was more active in BRAF mutated versus BRAF wild-type colon cancer cells., Conclusions: The cytotoxic effects of the copper thiosemicarbazone VLX60 differ from those of VLX50 and shows interesting features as a potential antitumor drug, notably against BRAF mutated colorectal cancer.
- Published
- 2017
- Full Text
- View/download PDF
30. In vitro and in vivo anti-leukemic activity of the peptidase-potentiated alkylator melflufen in acute myeloid leukemia.
- Author
-
Strese S, Hassan SB, Velander E, Haglund C, Höglund M, Larsson R, and Gullbo J
- Subjects
- Animals, Antigens, CD34 metabolism, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cytarabine pharmacology, Daunorubicin pharmacology, Dose-Response Relationship, Drug, Drug Synergism, Female, HL-60 Cells, Humans, Inhibitory Concentration 50, Leukemia, Myeloid, Acute enzymology, Leukemia, Myeloid, Acute pathology, Male, Melphalan pharmacology, Mice, SCID, Middle Aged, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Phenylalanine pharmacology, Time Factors, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents, Alkylating pharmacology, Leukemia, Myeloid, Acute drug therapy, Melphalan analogs & derivatives, Phenylalanine analogs & derivatives
- Abstract
The novel aminopeptidase potentiated alkylating agent melflufen, was evaluated for activity in acute myeloid leukemia in a range of in vitro models, as well as in a patient derived xenograft study. All tested AML cell lines were highly sensitive to melflufen while melphalan was considerably less potent. In the HL-60 cell line model, synergy was observed for the combination of melflufen and cytarabine, an interaction that appeared sequence dependent with increased synergy when melflufen was added before cytarabine. Also, in primary cultures of AML cells from patients melflufen was highly active, while normal PBMC cultures appeared less sensitive, indicating a 7-fold in vitro therapeutic index. Melphalan, on the other hand, was only 2-fold more potent in the AML patient samples compared with PBMCs. Melflufen was equally active against non-malignant, immature CD34+ progenitor cells and a more differentiated CD34+ derived cell population (GM14), whereas the stem cell like cells were less sensitive to melphalan. Finally, melflufen treatment showed significant anti-leukemia activity and increased survival in a patient derived xenograft of AML in mice. In conclusion, melflufen demonstrates high and significant preclinical activity in AML and further clinical evaluation seem warranted in this disease.
- Published
- 2017
- Full Text
- View/download PDF
31. Iron chelators target both proliferating and quiescent cancer cells.
- Author
-
Fryknäs M, Zhang X, Bremberg U, Senkowski W, Olofsson MH, Brandt P, Persson I, D'Arcy P, Gullbo J, Nygren P, Schughart LK, Linder S, and Larsson R
- Subjects
- Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Deferoxamine pharmacology, Dose-Response Relationship, Drug, HCT116 Cells, HT29 Cells, Humans, Hydrazones chemistry, Inhibitory Concentration 50, Iron Chelating Agents chemistry, MCF-7 Cells, Mitochondria metabolism, Ribonucleotide Reductases antagonists & inhibitors, Ribonucleotide Reductases metabolism, Structure-Activity Relationship, Triazoles chemistry, Antineoplastic Agents pharmacology, Cell Cycle drug effects, Hydrazones pharmacology, Iron Chelating Agents pharmacology, Mitochondria drug effects, Triazoles pharmacology
- Abstract
Poorly vascularized areas of solid tumors contain quiescent cell populations that are resistant to cell cycle-active cancer drugs. The compound VLX600 was recently identified to target quiescent tumor cells and to inhibit mitochondrial respiration. We here performed gene expression analysis in order to characterize the cellular response to VLX600. The compound-specific signature of VLX600 revealed a striking similarity to signatures generated by compounds known to chelate iron. Validation experiments including addition of ferrous and ferric iron in excess, EXAFS measurements, and structure activity relationship analyses showed that VLX600 chelates iron and supported the hypothesis that the biological effects of this compound is due to iron chelation. Compounds that chelate iron possess anti-cancer activity, an effect largely attributed to inhibition of ribonucleotide reductase in proliferating cells. Here we show that iron chelators decrease mitochondrial energy production, an effect poorly tolerated by metabolically stressed tumor cells. These pleiotropic features make iron chelators an attractive option for the treatment of solid tumors containing heterogeneous populations of proliferating and quiescent cells., Competing Interests: M.F., J.G., P.N., S.L. and R.L. are minor shareholders in Vivolux AB. The remaining authors have no competing financial interests.
- Published
- 2016
- Full Text
- View/download PDF
32. Preclinical activity of melflufen (J1) in ovarian cancer.
- Author
-
Carlier C, Strese S, Viktorsson K, Velander E, Nygren P, Uustalu M, Juntti T, Lewensohn R, Larsson R, Spira J, De Vlieghere E, Ceelen WP, and Gullbo J
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Line, Tumor, Cytoreduction Surgical Procedures, Disease-Free Survival, Drug Evaluation, Preclinical, Female, Humans, Hyperthermia, Induced, Injections, Intraperitoneal, Melphalan therapeutic use, Mice, Mice, SCID, Neoplasm Recurrence, Local, Neoplasm Staging, Ovarian Neoplasms pathology, Peritoneal Neoplasms pathology, Phenylalanine therapeutic use, Xenograft Model Antitumor Assays, Melphalan analogs & derivatives, Ovarian Neoplasms drug therapy, Peritoneal Neoplasms drug therapy, Phenylalanine analogs & derivatives
- Abstract
Ovarian cancer carries a significant mortality. Since symptoms tend to be minimal, the disease is often diagnosed when peritoneal metastases are already present. The standard of care in advanced ovarian cancer consists of platinum-based chemotherapy combined with cytoreductive surgery. Unfortunately, even after optimal cytoreduction and adjuvant chemotherapy, most patients with stage III disease will develop a recurrence. Intraperitoneal administration of chemotherapy is an alternative treatment for patients with localized disease. The pharmacological and physiochemical properties of melflufen, a peptidase potentiated alkylator, raised the hypothesis that this drug could be useful in ovarian cancer and particularily against peritoneal carcinomatosis. In this study the preclinical effects of melflufen were investigated in different ovarian cancer models. Melflufen was active against ovarian cancer cell lines, primary cultures of patient-derived ovarian cancer cells, and inhibited the growth of subcutaneous A2780 ovarian cancer xenografts alone and when combined with gemcitabine or liposomal doxorubicin when administered intravenously. In addition, an intra- and subperitoneal xenograft model showed activity of intraperitoneal administered melflufen for peritoneal carcinomatosis, with minimal side effects and modest systemic exposure. In conclusion, results from this study support further investigations of melflufen for the treatment of peritoneal carcinomatosis from ovarian cancer, both for intravenous and intraperitoneal administration.
- Published
- 2016
- Full Text
- View/download PDF
33. Corrigendum: The proteasome deubiquitinase inhibitor VLX1570 shows selectivity for ubiquitin-specific protease-14 and induces apoptosis of multiple myeloma cells.
- Author
-
Wang X, Mazurkiewicz M, Hillert EK, Olofsson MH, Pierrou S, Hillertz P, Gullbo J, Selvaraju K, Paulus A, Akhtar S, Bossler F, Khan AC, Linder S, and D'Arcy P
- Published
- 2016
- Full Text
- View/download PDF
34. The proteasome deubiquitinase inhibitor VLX1570 shows selectivity for ubiquitin-specific protease-14 and induces apoptosis of multiple myeloma cells.
- Author
-
Wang X, Mazurkiewicz M, Hillert EK, Olofsson MH, Pierrou S, Hillertz P, Gullbo J, Selvaraju K, Paulus A, Akhtar S, Bossler F, Khan AC, Linder S, and D'Arcy P
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents metabolism, Azepines chemistry, Azepines metabolism, Benzylidene Compounds chemistry, Benzylidene Compounds metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival, Enzyme Stability, Female, Humans, Mice, SCID, Polyubiquitin metabolism, Proteasome Endopeptidase Complex metabolism, Proteasome Inhibitors chemistry, Proteasome Inhibitors metabolism, Protein Binding, Proteolysis, Ubiquitin Thiolesterase chemistry, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Apoptosis drug effects, Azepines pharmacology, Benzylidene Compounds pharmacology, Multiple Myeloma drug therapy, Proteasome Inhibitors pharmacology, Ubiquitin Thiolesterase antagonists & inhibitors
- Abstract
Inhibition of deubiquitinase (DUB) activity is a promising strategy for cancer therapy. VLX1570 is an inhibitor of proteasome DUB activity currently in clinical trials for relapsed multiple myeloma. Here we show that VLX1570 binds to and inhibits the activity of ubiquitin-specific protease-14 (USP14) in vitro, with comparatively weaker inhibitory activity towards UCHL5 (ubiquitin-C-terminal hydrolase-5). Exposure of multiple myeloma cells to VLX1570 resulted in thermostabilization of USP14 at therapeutically relevant concentrations. Transient knockdown of USP14 or UCHL5 expression by electroporation of siRNA reduced the viability of multiple myeloma cells. Treatment of multiple myeloma cells with VLX1570 induced the accumulation of proteasome-bound high molecular weight polyubiquitin conjugates and an apoptotic response. Sensitivity to VLX1570 was moderately affected by altered drug uptake, but was unaffected by overexpression of BCL2-family proteins or inhibitors of caspase activity. Finally, treatment with VLX1570 was found to lead to extended survival in xenograft models of multiple myeloma. Our findings demonstrate promising antiproliferative activity of VLX1570 in multiple myeloma, primarily associated with inhibition of USP14 activity.
- Published
- 2016
- Full Text
- View/download PDF
35. In vitro and in vivo activity of melflufen (J1)in lymphoma.
- Author
-
Delforoush M, Strese S, Wickström M, Larsson R, Enblad G, and Gullbo J
- Subjects
- Animals, Cell Line, Tumor, G2 Phase Cell Cycle Checkpoints drug effects, Humans, Lymphoma pathology, Melphalan administration & dosage, Melphalan adverse effects, Mice, Multiple Myeloma pathology, Phenylalanine administration & dosage, Phenylalanine adverse effects, Xenograft Model Antitumor Assays, Cell Proliferation drug effects, Lymphoma drug therapy, Melphalan analogs & derivatives, Multiple Myeloma drug therapy, Phenylalanine analogs & derivatives
- Abstract
Background: Melphalan has been used in the treatment of various hematologic malignancies for almost 60 years. Today it is part of standard therapy for multiple myeloma and also as part of myeloablative regimens in association with autologous allogenic stem cell transplantation. Melflufen (melphalan flufenamide ethyl ester, previously called J1) is an optimized derivative of melphalan providing targeted delivery of active metabolites to cells expressing aminopeptidases. The activity of melflufen has compared favorably with that of melphalan in a series of in vitro and in vivo experiments performed preferentially on different solid tumor models and multiple myeloma. Melflufen is currently being evaluated in a clinical phase I/II trial in relapsed or relapsed and refractory multiple myeloma., Methods: Cytotoxicity of melflufen was assayed in lymphoma cell lines and in primary tumor cells with the Fluorometric Microculture Cytotoxicity Assay and cell cycle analyses was performed in two of the cell lines. Melflufen was also investigated in a xenograft model with subcutaneous lymphoma cells inoculated in mice., Results: Melflufen showed activity with cytotoxic IC50-values in the submicromolar range (0.011-0.92 μM) in the cell lines, corresponding to a mean of 49-fold superiority (p < 0.001) in potency vs. melphalan. In the primary cultures melflufen yielded slightly lower IC50-values (2.7 nM to 0.55 μM) and an increased ratio vs. melphalan (range 13-455, average 108, p < 0.001). Treated cell lines exhibited a clear accumulation in the G2/M-phase of the cell cycle. Melflufen also showed significant activity and no, or minimal side effects in the xenografted animals., Conclusion: This study confirms previous reports of a targeting related potency superiority of melflufen compared to that of melphalan. Melflufen was active in cell lines and primary cultures of lymphoma cells, as well as in a xenograft model in mice and appears to be a candidate for further evaluation in the treatment of this group of malignant diseases.
- Published
- 2016
- Full Text
- View/download PDF
36. Targeting Mitochondrial Function to Treat Quiescent Tumor Cells in Solid Tumors.
- Author
-
Zhang X, de Milito A, Olofsson MH, Gullbo J, D'Arcy P, and Linder S
- Subjects
- Animals, Cell Proliferation drug effects, Drug Discovery, Drug Resistance, Neoplasm, Drug Screening Assays, Antitumor, Energy Metabolism drug effects, Glucose metabolism, Humans, Hypoxia, Molecular Targeted Therapy, Neoplasms pathology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Oxidative Phosphorylation drug effects, Spheroids, Cellular drug effects, Spheroids, Cellular metabolism, Tumor Cells, Cultured, Tumor Microenvironment drug effects, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Mitochondria drug effects, Mitochondria metabolism, Neoplasms drug therapy, Neoplasms metabolism, Resting Phase, Cell Cycle drug effects
- Abstract
The disorganized nature of tumor vasculature results in the generation of microenvironments characterized by nutrient starvation, hypoxia and accumulation of acidic metabolites. Tumor cell populations in such areas are often slowly proliferating and thus refractory to chemotherapeutical drugs that are dependent on an active cell cycle. There is an urgent need for alternative therapeutic interventions that circumvent growth dependency. The screening of drug libraries using multicellular tumor spheroids (MCTS) or glucose-starved tumor cells has led to the identification of several compounds with promising therapeutic potential and that display activity on quiescent tumor cells. Interestingly, a common theme of these drug screens is the recurrent identification of agents that affect mitochondrial function. Such data suggest that, contrary to the classical Warburg view, tumor cells in nutritionally-compromised microenvironments are dependent on mitochondrial function for energy metabolism and survival. These findings suggest that mitochondria may represent an "Achilles heel" for the survival of slowly-proliferating tumor cells and suggest strategies for the development of therapy to target these cell populations.
- Published
- 2015
- Full Text
- View/download PDF
37. Induction of mitochondrial dysfunction as a strategy for targeting tumour cells in metabolically compromised microenvironments.
- Author
-
Zhang X, Fryknäs M, Hernlund E, Fayad W, De Milito A, Olofsson MH, Gogvadze V, Dang L, Påhlman S, Schughart LA, Rickardson L, D'Arcy P, Gullbo J, Nygren P, Larsson R, and Linder S
- Subjects
- Animals, Autophagy drug effects, Female, Glucose metabolism, Glycolysis drug effects, HCT116 Cells, HT29 Cells, Humans, Hypoxia chemically induced, Mice, Mitochondria metabolism, Oxidative Phosphorylation drug effects, Spheroids, Cellular, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Screening Assays, Antitumor, Hydrazones pharmacology, Mitochondria drug effects, Triazoles pharmacology, Tumor Microenvironment
- Abstract
Abnormal vascularization of solid tumours results in the development of microenvironments deprived of oxygen and nutrients that harbour slowly growing and metabolically stressed cells. Such cells display enhanced resistance to standard chemotherapeutic agents and repopulate tumours after therapy. Here we identify the small molecule VLX600 as a drug that is preferentially active against quiescent cells in colon cancer 3-D microtissues. The anticancer activity is associated with reduced mitochondrial respiration, leading to bioenergetic catastrophe and tumour cell death. VLX600 shows enhanced cytotoxic activity under conditions of nutrient starvation. Importantly, VLX600 displays tumour growth inhibition in vivo. Our findings suggest that tumour cells in metabolically compromised microenvironments have a limited ability to respond to decreased mitochondrial function, and suggest a strategy for targeting the quiescent populations of tumour cells for improved cancer treatment.
- Published
- 2014
- Full Text
- View/download PDF
38. Screening for phenotype selective activity in multidrug resistant cells identifies a novel tubulin active agent insensitive to common forms of cancer drug resistance.
- Author
-
Fryknäs M, Gullbo J, Wang X, Rickardson L, Jarvius M, Wickström M, Hassan S, Andersson C, Gustafsson M, Westman G, Nygren P, Linder S, and Larsson R
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Drug Resistance, Multiple, Drug Screening Assays, Antitumor, Flow Cytometry, Inhibitory Concentration 50, Mice, Oligonucleotide Array Sequence Analysis, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm, Hydroxyquinolines pharmacology, Neoplasms, Tubulin drug effects
- Abstract
Background: Drug resistance is a common cause of treatment failure in cancer patients and encompasses a multitude of different mechanisms. The aim of the present study was to identify drugs effective on multidrug resistant cells., Methods: The RPMI 8226 myeloma cell line and its multidrug resistant subline 8226/Dox40 was screened for cytotoxicity in response to 3,000 chemically diverse compounds using a fluorometric cytotoxicity assay (FMCA). Follow-up profiling was subsequently performed using various cellular and biochemical assays., Results: One compound, designated VLX40, demonstrated a higher activity against 8226/Dox40 cells compared to its parental counterpart. VLX40 induced delayed cell death with apoptotic features. Mechanistic exploration was performed using gene expression analysis of drug exposed tumor cells to generate a drug-specific signature. Strong connections to tubulin inhibitors and microtubule cytoskeleton were retrieved. The mechanistic hypothesis of VLX40 acting as a tubulin inhibitor was confirmed by direct measurements of interaction with tubulin polymerization using a biochemical assay and supported by demonstration of G2/M cell cycle arrest. When tested against a broad panel of primary cultures of patient tumor cells (PCPTC) representing different forms of leukemia and solid tumors, VLX40 displayed high activity against both myeloid and lymphoid leukemias in contrast to the reference compound vincristine to which myeloid blast cells are often insensitive. Significant in vivo activity was confirmed in myeloid U-937 cells implanted subcutaneously in mice using the hollow fiber model., Conclusions: The results indicate that VLX40 may be a useful prototype for development of novel tubulin active agents that are insensitive to common mechanisms of cancer drug resistance.
- Published
- 2013
- Full Text
- View/download PDF
39. Alternative cytotoxic effects of the postulated IGF-IR inhibitor picropodophyllin in vitro.
- Author
-
Wu X, Sooman L, Wickström M, Fryknäs M, Dyrager C, Lennartsson J, and Gullbo J
- Subjects
- Carcinoma, Squamous Cell metabolism, Cell Line, Tumor, Dose-Response Relationship, Drug, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Esophageal Neoplasms metabolism, Humans, Microtubules metabolism, Phosphorylation drug effects, Podophyllotoxin toxicity, Protein Kinase Inhibitors pharmacology, Receptor, IGF Type 1 metabolism, Signal Transduction drug effects, Tubulin Modulators pharmacology, Antineoplastic Agents toxicity, Podophyllotoxin analogs & derivatives, Receptor, IGF Type 1 antagonists & inhibitors
- Abstract
The insulin-like growth factor-1 (IGF-I) and its receptors play an important role in transformation and progression of several malignancies. Inhibitors of this pathway have been developed and evaluated but generally performed poorly in clinical trials, and several drug candidates have been abandoned. The cyclolignan picropodophyllin (PPP) has been described as a potent and selective IGF-IR inhibitor and is currently undergoing clinical trials. We investigated PPP's activity in panels of human cancer cell lines (e.g., esophageal squamous carcinoma cell lines) but found no effects on the phosphorylation or expression of IGF-IR. Nor was the cytotoxic activity of PPP related to the presence or spontaneous phosphorylation of IGF-IR. However, its activity correlated with that of known tubulin inhibitors, and it destabilized microtubule assembly at cytotoxic concentrations also achievable in patients. PPP is a stereoisomer of podophyllotoxin (PPT), a potent tubulin inhibitor, and an equilibrium between the two has previously been described. PPP could thus potentially act as a reservoir for the continuous generation of low doses of PPT. Interestingly, PPP also inhibited downstream signaling from tyrosine kinase receptors, including the serine/threonine kinase Akt. This effect is associated with microtubule-related downregulation of the EGF receptor, rather than the IGF-IR. These results suggest that the cytotoxicity and pAkt inhibition observed following treatment with the cyclolignan PPP in vitro result from microtubule inhibition (directly or indirectly by spontaneous PPT formation), rather than any effect on IGF-IR. It is also suggested that PPT should be used as a reference compound in all future studies on PPP.
- Published
- 2013
- Full Text
- View/download PDF
40. Effects of hypoxia on human cancer cell line chemosensitivity.
- Author
-
Strese S, Fryknäs M, Larsson R, and Gullbo J
- Subjects
- Cell Line, Tumor, Cell Survival drug effects, Drug Screening Assays, Antitumor, Humans, Antineoplastic Agents pharmacology, Cell Hypoxia, Cell Proliferation drug effects, Drug Resistance, Neoplasm physiology, Neoplasms metabolism
- Abstract
Background: Environment inside even a small tumor is characterized by total (anoxia) or partial oxygen deprivation, (hypoxia). It has been shown that radiotherapy and some conventional chemotherapies may be less effective in hypoxia, and therefore it is important to investigate how different drugs act in different microenvironments. In this study we perform a large screening of the effects of 19 clinically used or experimental chemotherapeutic drugs on five different cell lines in conditions of normoxia, hypoxia and anoxia., Methods: A panel of 19 commercially available drugs: 5-fluorouracil, acriflavine, bortezomib, cisplatin, digitoxin, digoxin, docetaxel, doxorubicin, etoposide, gemcitabine, irinotecan, melphalan, mitomycin c, rapamycin, sorafenib, thalidomide, tirapazamine, topotecan and vincristine were tested for cytotoxic activity on the cancer cell lines A2780 (ovarian), ACHN (renal), MCF-7 (breast), H69 (SCLC) and U-937 (lymphoma). Parallel aliquots of the cells were grown at different oxygen pressures and after 72 hours of drug exposure viability was measured with the fluorometric microculture cytotoxicity assay (FMCA)., Results: Sorafenib, irinotecan and docetaxel were in general more effective in an oxygenated environment, while cisplatin, mitomycin c and tirapazamine were more effective in a low oxygen environment. Surprisingly, hypoxia in H69 and MCF-7 cells mostly rendered higher drug sensitivity. In contrast ACHN appeared more sensitive to hypoxia, giving slower proliferating cells, and consequently, was more resistant to most drugs., Conclusions: A panel of standard cytotoxic agents was tested against five different human cancer cell lines cultivated at normoxic, hypoxic and anoxic conditions. Results show that impaired chemosensitivity is not universal, in contrast different cell lines behave different and some drugs appear even less effective in normoxia than hypoxia.
- Published
- 2013
- Full Text
- View/download PDF
41. The Nanoparticulate Quillaja Saponin BBE is selectively active towards renal cell carcinoma.
- Author
-
Hassan SB, Gullbo J, Hu K, Berenjian S, Morein B, and Nygren P
- Subjects
- Animals, Apoptosis drug effects, Carcinoma, Renal Cell drug therapy, Cell Line, Tumor, Drug Resistance, Neoplasm, Female, Humans, Kidney Neoplasms drug therapy, Mice, Mice, Nude, Nanoparticles administration & dosage, Transplantation, Heterologous, Cell Proliferation drug effects, Plant Extracts administration & dosage, Plant Extracts chemistry, Quillaja chemistry, Quillaja Saponins administration & dosage
- Abstract
Aim: To characterize the cytotoxic effect of BBE, the particulate of desacyl-saponin, in model systems of solid tumours., Materials and Methods: Cytotoxic activity of BBE was investigated in solid human tumour cell lines, in tumour cells from patients with renal cell carcinoma, in normal human renal cells and in peripheral blood mononuclear cells. The BBE mode of cell death was assessed in vitro. In vivo effect of BBE was evaluated in xenograft-bearing mice., Results: BBE was selectively active against renal cell carcinoma, with no or little effect on normal cells. BBE induced caspase activity and apoptosis. An inhibitory activity of BBE on xenograft tumour growth, with no apparent signs of haematological toxicity was shown. In the non-proliferative model of patient tumour cells, BBE was active on only 1/5 patient samples, suggesting association of BBE effect with cell proliferation., Conclusion: BBE has interesting activities against renal cell carcinoma and should be further explored as a drug against this resistant tumour type.
- Published
- 2013
42. Novel activity of acriflavine against colorectal cancer tumor cells.
- Author
-
Hassan S, Laryea D, Mahteme H, Felth J, Fryknäs M, Fayad W, Linder S, Rickardson L, Gullbo J, Graf W, Påhlman L, Glimelius B, Larsson R, and Nygren P
- Subjects
- Camptothecin analogs & derivatives, Camptothecin pharmacology, Cell Line, Tumor drug effects, DNA Topoisomerases, Type I metabolism, DNA Topoisomerases, Type II metabolism, Drug Screening Assays, Antitumor, Female, Fluorouracil pharmacology, High-Throughput Screening Assays, Humans, Irinotecan, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Male, Organoplatinum Compounds pharmacology, Ovarian Neoplasms drug therapy, Oxaliplatin, Acriflavine pharmacology, Colorectal Neoplasms drug therapy, Topoisomerase I Inhibitors pharmacology, Topoisomerase II Inhibitors pharmacology
- Abstract
A high-throughput screen of the cytotoxic activity of 2000 molecules from a commercial library in three human colon cancer cell lines and two normal cell types identified the acridine acriflavin to be a colorectal cancer (CRC) active drug. Acriflavine was active in cell spheroids, indicating good drug penetration and activity against hypoxic cells. In a validation step based on primary cultures of patient tumor cells, acriflavine was found to be more active against CRC than ovarian cancer and chronic lymphocytic leukemia. This contrasted to the activity pattern of the CRC active standard drugs 5-fluorouracil, irinotecan and oxaliplatin. Mechanistic studies indicated acriflavine to be a dual topoisomerase I and II inhibitor. In conclusion, the strategy used seems promising for identification of new diagnosis-specific cancer drugs., (© 2011 Japanese Cancer Association.)
- Published
- 2011
- Full Text
- View/download PDF
43. Aminopeptidase N (CD13) as a target for cancer chemotherapy.
- Author
-
Wickström M, Larsson R, Nygren P, and Gullbo J
- Subjects
- CD13 Antigens analysis, CD13 Antigens physiology, Cell Proliferation, Clinical Trials as Topic, Glycine analogs & derivatives, Glycine therapeutic use, Humans, Hydroxamic Acids therapeutic use, Leucine analogs & derivatives, Leucine therapeutic use, Neoplasm Invasiveness, Neovascularization, Pathologic etiology, Oligopeptides administration & dosage, Prognosis, CD13 Antigens antagonists & inhibitors, Neoplasms drug therapy, Protease Inhibitors therapeutic use
- Abstract
The enzyme aminopeptidase N (APN, also known as CD13) is a Zn(2+) dependent membrane-bound ectopeptidase that degrades preferentially proteins and peptides with a N-terminal neutral amino acid. Aminopeptidase N has been associated with the growth of different human cancers and suggested as a suitable target for anti-cancerous therapy. Different approaches have been used to develop new drugs directed to this target, including enzyme inhibitors as well as APN-targeted carrier constructs. This review discusses the prevalence and possible function of APN in malignant diseases, mainly solid tumors, as well as its "drugability" evaluated in preclinical in vivo models, and also provides a brief overview of current clinical trials focused on APN., (© 2011 Japanese Cancer Association.)
- Published
- 2011
- Full Text
- View/download PDF
44. Ex vivo activity of cardiac glycosides in acute leukaemia.
- Author
-
Hallböök H, Felth J, Eriksson A, Fryknäs M, Bohlin L, Larsson R, and Gullbo J
- Subjects
- Acute Disease, Cardiac Glycosides therapeutic use, Cell Line, Tumor, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Digitoxin pharmacology, Humans, Inhibitory Concentration 50, Leukemia pathology, Leukemia, B-Cell drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Biosynthesis drug effects, Species Specificity, Cardiac Glycosides pharmacology, Leukemia drug therapy
- Abstract
Background: Despite years of interest in the anti-cancerous effects of cardiac glycosides (CGs), and numerous studies in vitro and in animals, it has not yet been possible to utilize this potential clinically. Reports have demonstrated promising in vitro effects on different targets as well as a possible therapeutic index/selectivity in vitro and in experimental animals. Recently, however, general inhibition of protein synthesis was suggested as the main mechanism of the anti-cancerous effects of CGs. In addition, evidence of species differences of a magnitude sufficient to explain the results of many studies called for reconsideration of earlier results., Principal Findings: In this report we identified primary B-precursor and T-ALL cells as being particularly susceptible to the cytotoxic effects of CGs. Digitoxin appeared most potent and IC(50) values for several patient samples were at concentrations that may be achieved in the clinic. Significant protein synthesis inhibition at concentrations corresponding to IC(50) was demonstrated in colorectal tumour cell lines moderately resistant to the cytotoxic effects of digoxin and digitoxin, but not in highly sensitive leukaemia cell lines., Conclusion: It is suggested that further investigation regarding CGs may be focused on diagnoses like T- and B-precursor ALL.
- Published
- 2011
- Full Text
- View/download PDF
45. Nanoparticulate Quillaja saponin induces apoptosis in human leukemia cell lines with a high therapeutic index.
- Author
-
Hu K, Berenjian S, Larsson R, Gullbo J, Nygren P, Lövgren T, and Morein B
- Subjects
- Antineoplastic Agents administration & dosage, Antineoplastic Agents chemistry, Drug Compounding methods, Humans, Jurkat Cells, Particle Size, Treatment Outcome, U937 Cells, Apoptosis drug effects, Nanoparticles administration & dosage, Nanoparticles chemistry, Phytotherapy methods, Plant Extracts administration & dosage, Plant Extracts chemistry, Quillaja chemistry
- Abstract
Saponin fractions of Quillaja saponaria Molina (QS) have cytotoxic activity against cancer cells in vitro, but are too toxic to be useful in the clinic. The toxic effect was abolished by converting QS fractions into stable nanoparticles through the binding of QS to cholesterol. Two fractions of QS were selected for particle formation, one with an acyl-chain (ASAP) was used to form killing and growth-inhibiting (KGI) particles, and the other without the acyl-chain (DSAP) was used to formulate blocking and balancing effect (BBE) particles. KGI showed significant growth inhibiting and cancer cell-killing activities in nine of 10 cell lines while BBE showed that on one cell line. The monoblastoid lymphoma cell line U937 was selected for analyzing the mode of action. Low concentrations of KGI (0.5 and 2 microg/mL) induced irreversible exit from the cell cycle, differentiation measured by cytokine production, and eventually programmed cell death (apoptosis). Compared to normal human monocytes, the U937 cells were 30-fold more sensitive to KGI. The nontoxic BBE blocked the cell killing effect of KGI in a concentration-dependent manner. In conclusion, the formulation of QS into nanoparticles has the potential of becoming a new class of anticancer agents.
- Published
- 2010
46. Structure-activity relationship analysis of cytotoxic cyanoguanidines: selection of CHS 828 as candidate drug.
- Author
-
Lövborg H, Burman R, and Gullbo J
- Abstract
Background: N-(6-(4-chlorophenoxy)hexyl)-N'-cyano-N''-4-pyridyl guanidine) (CHS 828) is the first candidate drug from a novel group of anti-tumour agents - the pyridyl cyanoguanidines, shown to be potent compounds interfering with cellular metabolism (inhibition of nicotinamide phosphoribosyl transferase) and NF-kappaB signalling. Substituted cyanoguanidines are also found in anti-hypertensive agents such as the potassium channel opener pinacidil (N-cyano-N'-(4-pyridyl)-N''-(1,2,2-trimethylpropyl)guanidine) and histamine-II receptor antagonists (e.g. cimetidine, N-cyano-N'-methyl-N''-[2-[[(5-methylimidazol-4-yl]methyl]thio]ethyl)guanidine). In animal studies, CHS 828 has shown very promising activity, and phase I and II studies resulted in further development of a with a water soluble prodrug., Findings: To study the structural requirements for cyanoguanidine cytotoxicity a set of 19 analogues were synthesized. The cytotoxic effects were then studied in ten cell lines selected for different origins and mechanisms of resistance, using the fluorometric microculture cytotoxicity assay (FMCA). The compounds showed varying cytotoxic activity even though the dose-response curves for some analogues were very shallow. Pinacidil and cimetidine were found to be non-toxic in all ten cell lines. Starting with cyanoguanidine as the crucial core it was shown that 4-pyridyl substitution was more efficient than was 3-pyridyl substitution. The 4-pyridyl cyanoguanidine moiety should be linked by an alkyl chain, optimally a hexyl, heptyl or octyl chain, to a bulky end group. The exact composition of this end group did not seem to be of crucial importance; when the end group was a mono-substituted phenyl ring it was shown that the preferred position was 4-substitution, followed by 3- and, finally, 2-substitution as the least active. Whether the substituent was a chloro, nitro or methoxy substituent seemed to be of minor importance. Finally, the activity patterns in the ten cell lines were compared. Substances with similar structures correlated well, whilst substances with large differences in molecular structure demonstrated lower correlation coefficients., Conclusion: According to this structure-activity relationship (SAR) study, CHS 828 meets the requirements for optimal cytotoxic activity for this class of compounds.
- Published
- 2009
- Full Text
- View/download PDF
47. The novel melphalan prodrug J1 inhibits neuroblastoma growth in vitro and in vivo.
- Author
-
Wickström M, Johnsen JI, Ponthan F, Segerström L, Sveinbjörnsson B, Lindskog M, Lövborg H, Viktorsson K, Lewensohn R, Kogner P, Larsson R, and Gullbo J
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Apoptosis drug effects, Carboplatin administration & dosage, Cell Line, Tumor drug effects, Cell Survival drug effects, Dose-Response Relationship, Drug, Doxorubicin administration & dosage, Drug Screening Assays, Antitumor, Etoposide administration & dosage, Female, Humans, Immunoenzyme Techniques, Male, Melphalan administration & dosage, Mice, Molecular Structure, Neovascularization, Pathologic, Neuroblastoma blood supply, Neuroblastoma metabolism, Rats, Rats, Nude, Tumor Cells, Cultured, Vincristine administration & dosage, Dipeptides pharmacology, Neuroblastoma drug therapy, Prodrugs pharmacology
- Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood. The activity of J1 (l-melphalanyl-p-l-fluorophenylalanine ethyl ester), an enzymatically activated melphalan prodrug, was evaluated in neuroblastoma models in vitro and in vivo. Seven neuroblastoma cell lines with various levels of drug resistance were screened for cytotoxicity of J1 alone or in combination with standard cytotoxic drugs, using a fluorometric cytotoxicity assay. J1 displayed high cytotoxic activity in vitro against all neuroblastoma cell lines, with IC(50) values in the submicromolar range, significantly more potent than melphalan. The cytotoxicity of J1, but not melphalan, could be significantly inhibited by the aminopeptidase inhibitor bestatin. J1 induced caspase-3 cleavage and apoptotic morphology, had additive effects in combination with doxorubicin, cyclophosphamide, carboplatin, and vincristine, and synergistically killed otherwise drug-resistant cells when combined with etoposide. Athymic rats and mice carrying neuroblastoma xenografts [SH-SY5Y, SK-N-BE(2)] were treated with equimolar doses of melphalan, J1, or no drug, and effects on tumor growth and tissue morphology were analyzed. Tumor growth in vivo was significantly inhibited by J1 compared with untreated controls. Compared with melphalan, J1 more effectively inhibited the growth of mice with SH-SY5Y xenografts, was associated with higher caspase-3 activation, fewer proliferating tumor cells, and significantly decreased mean vascular density. In conclusion, the melphalan prodrug J1 is highly active in models of neuroblastoma in vitro and in vivo, encouraging further clinical development in this patient group.
- Published
- 2007
- Full Text
- View/download PDF
48. Clinical value of using serological cytokeratins as therapeutic markers in thoracic malignancies.
- Author
-
Ekman S, Eriksson P, Bergström S, Johansson P, Goike H, Gullbo J, Henriksson R, Larsson A, and Bergqvist M
- Subjects
- Biological Assay, Humans, Biomarkers, Tumor blood, Keratins blood, Thoracic Neoplasms blood, Thoracic Neoplasms diagnosis
- Abstract
In recent years, there has been an increasing awareness among physicians of the value of therapeutic interventions in patients suffering from lung cancer and mesothelioma. A search for an optimal approach using surgery, irradiation and chemotherapy in different settings of the tumour disease, including curatively aimed adjuvant chemotherapy after locoregional surgery or radiotherapy, has resulted in gradually improved survival rates. Still, early detection is crucial if there is to be a possibility of curing patients or prolonging life in cases of relapsed disease. Several studies have been initiated in which surrogate markers are evaluated in comparison to chest X-rays and computer tomography. The present review focuses on the predictive and prognostic value of using serological cytokeratins as tumour markers for patients suffering from thoracic malignancies.
- Published
- 2007
49. Phenotype-based screening of mechanistically annotated compounds in combination with gene expression and pathway analysis identifies candidate drug targets in a human squamous carcinoma cell model.
- Author
-
Fryknäs M, Rickardson L, Wickström M, Dhar S, Lövborg H, Gullbo J, Nygren P, Gustafsson MG, Isaksson A, and Larsson R
- Subjects
- Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell genetics, Caspases metabolism, Combinatorial Chemistry Techniques methods, Drug Evaluation, Preclinical methods, HeLa Cells, Humans, Models, Biological, Oligonucleotide Array Sequence Analysis, Phenotype, Phosphodiesterase Inhibitors pharmacology, Phosphoric Diester Hydrolases metabolism, Pyridazines pharmacology, Quinazolines, RNA, Messenger metabolism, Carcinoma, Squamous Cell metabolism, Electronic Data Processing methods, Gene Expression Profiling methods, Signal Transduction genetics
- Abstract
The squamous cell carcinoma HeLa cell line and an epithelial cell line hTERT-RPE with a nonmalignant phenotype were interrogated for HeLa cell selectivity in response to 1267 annotated compounds representing 56 pharmacological classes. Selective cytotoxic activity was observed for 14 of these compounds dominated by cyclic adenosine monophosphate (cAMP) selective phosphodiesterase (PDE) inhibitors, which tended to span a representation of the chemical descriptor space of the library. The PDE inhibitors induced delayed cell death with features compatible with classical apoptosis. The PDE inhibitors were largely inactive when tested against a cell line panel consisting of hematological and nonsquamous epithelial phenotypes. In a genome-wide DNA microarray analysis, PDE3A and PDE2A were found to be significantly increased in HeLa cells compared to the other cell lines. The pathway analysis software PathwayAssist was subsequently used to extract a list of proteins and small molecules retrieved from Medline abstracts associated with the hit compounds. The resulting list consisted of major parts of the cAMP-protein kinase A pathway linking to ERK, P38, and AKT. This molecular network may provide a basis for further exploitation of novel candidate targets for the treatment of squamous cell carcinoma.
- Published
- 2006
- Full Text
- View/download PDF
50. Antitumor activity of the novel melphalan containing tripeptide J3 (L-prolyl-L-melphalanyl-p-L-fluorophenylalanine ethyl ester): comparison with its m-L-sarcolysin analogue P2.
- Author
-
Gullbo J, Wallinder C, Tullberg M, Lövborg H, Ehrsson H, Lewensohn R, Nygren P, Luthman K, and Larsson R
- Subjects
- Cell Line, Tumor, Dipeptides, Drug Screening Assays, Antitumor, Humans, Magnetic Resonance Spectroscopy, Antineoplastic Agents, Alkylating pharmacology, Melphalan analogs & derivatives, Melphalan pharmacology, Oligopeptides pharmacology
- Abstract
Peptichemio (PTC), a mixture of six oligopeptides all containing m-L-sarcolysin, has previously shown impressive results in clinical trials. The tripeptide P2 (L-prolyl-m-L-sarcolysyl-p-L-fluorophenylalanine ethyl ester) has been suggested as the main contributor to PTC activity. In contrast to its analogue melphalan, m-L-sarcolysin never reached clinical use. To allow a direct comparison, the corresponding melphalan containing tripeptide J3 (L-prolyl-L-melphalanyl-p-L-fluorophenylalanine ethyl ester) was synthesized and its activity was compared with that of P2; the activities of melphalan and m-L-sarcolysin were studied in parallel. Cytotoxic activity in human tumor cell lines and some fresh human tumor specimens were analyzed as well as effects on cellular metabolism, macromolecular synthesis, and preliminary evaluation of the cell death characteristics. The results show that melphalan and m-L-sarcolysin display similar activity in these systems and that the tripeptides were more active than their parent monomers. Surprisingly however, the melphalan containing tripeptide J3 demonstrated a significantly more rapid and stronger activity than the m-L-sarcolysin analogue P2. Finally, the in vivo toxicity and activity of melphalan and J3 were investigated in mice bearing human leukemia cells in s.c. fibers. The in vitro results seem translatable into the in vivo situation, demonstrating better antileukemic effect of J3 but similar side effects as melphalan.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.