eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2012
vol. 16
 
Share:
Share:
Original paper

Bortezomib has little ex vivo activity in chronic myeloid leukemia: individual tumor response testing comparative study in acute and chronic myeloid leukemia

Jan Styczyński
,
Lidia Gil
,
Krzysztof Czyżewski
,
Beata Kołodziej
,
Beata Kuryło-Rafińska
,
Krzysztof Lewandowski
,
Michał Gniot
,
Maria Lewandowska
,
Mieczysław Komarnicki
,
Mariusz Wysocki

Wspolczesna Onkol 2012; 16 (3): 210–214
Online publish date: 2012/07/06
Article file
- Bortezomib.pdf  [0.10 MB]
Get citation
 
PlumX metrics:
 

Background

Drug resistance is one of the factors contributing to poor response to therapy. Cellular drug resistance can be defined as a lack of cytotoxic response in cancer cells after administration of a cytotoxic compound. Response of cancer cells to chemotherapy can be tested in ex vivo conditions by several assays, such as the methylthiazol tetrazolium (MTT) assay, differential staining cytotoxicity (DiSC) assay, the fluorometric microculture cytotoxicity assay (FMCA) and similar assays. Considerable work based on these assays has been reported during the past 25 years, and recently an ad hoc group of 50 scientists from 10 countries agreed on the term “individualized tumor response (ITRT)” for these tests, describing them as the “effect of anticancer treatments on whole living tumor cells freshly removed from cancer patients” and not including tests with “subcellular fractions, animals or cell lines” [1, 2]. ITRT is regarded as an important risk factor of treatment failure in pediatric acute lymphoblastic leukemia (ALL). It can be demonstrated clinically as a poor steroid response after one-week monotherapy or as a delayed response of bone marrow at day +15 or day +33 of induction therapy. Presence of minimal residual disease also results in drug resistance. In comparison to pediatric ALL, the value of ITRT assays is less established in other types of leukemia, especially in chronic myeloid leukemia (CML). Introduction of tyrosine kinase inhibitors (TKIs) in therapy of CML has contributed to development of ex vivo testing in this disease. So far only very limited data on cellular drug resistance in CML cells are available [3–6].

The objective of the study was to analyze the ex vivo drug resistance profile to bortezomib and 22 other antileukemic drugs, including three tyrosine kinase inhibitors (TKIs), in CML in comparison to acute myeloid leukemia (AML).

Material and methods

Patients



A total of 82 patients entered the study, including 36 CML and 46 AML adults (age 18–69, median 41 years). However, due to technical reasons, not all drugs were tested for all patients. AML patients were diagnosed for de novo

(n = 20) or relapsed (n = 26) disease. CML patients were divided into the following subgroups: with advanced (n = 19) or non-advanced (n = 17) disease; with good (n = 20) or poor clinical response to imatinib

(n = 16) [7]; with (n = 6) or without mutation (n = 28). Non-advanced disease was defined as the first chronic CML phase. All other phases were classified as advanced disease. Poor clinical response was defined as clinical resistance to imatinib. All patients with a poor clinical response were tested for ABL-kinase domain mutations. Among CML patients,

19 had advanced disease, 16 were resistant to imatinib, and 6 had ABL-kinase domain mutations (M244V, E255K, Y253H, M351T and 2 with F317L).



Drugs



The following 20 drugs were used: bortezomib (Velcade, Janssen Pharmaceutica N.V., Beerse, Belgium; concentrations tested: 0.00019–2 M), prednisolone (Jelfa, Jelenia Góra, Poland; 0.0076–250 g/ml), vincristine (Gedeon Richter, Budapest, Hungary; 0.019–20 g/ml), L-asparaginase (Medac, Hamburg, Germany; 0.0032–10 IU/ml), daunorubicin (Rhone-Poulenc Rorer, Paris, France; 0.0019–2 g/ml), dox-

orubicin (Pharmacia Italia S.p.A., Milan, Italy; 0.031–40 g/mlz,

cytarabine (Upjohn, Puurs, Belgium; 0.24–250 g/ml), cladribine (Bioton, Warsaw, Poland; 0.0004–40 g/ml), etoposide (Bristol-Myers Squibb, Sermoneta, Italy; 0.048–

50 g/ml), thiotepa (Lederle, Wolfratshausen, Germany; 0.032–100 g/ml), topotecan (Glaxo SmithKline Manu-facturing S.p.A., Parma, Italy; 0.097–100 g/ml), busulfan (Busilvex, Pierre-Fabre-Medi­cament, Castres, France; 1.17 –1200 g/ml), 4-HOO-cyclophos­phamide (Asta Medica, Hamburg, Germany; 0.096–100 g/ml), fludarabine phosphate (Schering AG, Berlin, Germany; 0.019–20 g/ml), idarubicin (Pharmacia, Milan, Italy; 0.0019–2 g/ml), melphalan (Glaxo Wellcome, Parma, Italy; 0.038–40 g/ml), mitoxantrone (Jelfa; 0.001–1 g/ml), 6-thioguanine (Sigma, nr A4882; 1.56–50 g/ml), treosulfan (Medac; 0.0005–1 g/ml), and clofarabine (Bioenvsion / Genzyme, 0.01–12.5 M). Before the assay was carried out, most drug stock solutions were stored frozen in small aliquots at –20°C, except cladribine, which was stored at +4°C. Stock solutions were prepared in water for injection, and further dilution was made in respective medium.

CML patients were also tested for sensitivity to tyrosine kinase inhibitors: imatinib (Novartis Pharmaceuticals; concentrations tested: 0.000977–1 M), dasatinib (Bristol Meyers Squibb; 0.000977–1 M) and nilotinib (Novartis Pharmaceuticals; 0.000977–1 M).



Methylthiazol tetrazolium assay



Ex vivo drug resistance profile (ITRT) was studied by the MTT assay. The procedure of the assay is described elsewhere [2]. The concentration of drug that was lethal to 50% of the cells (LC50) was calculated from the dose response curve and was used as a measure for ex vivo drug resistance in each sample. Relative resistance (RR) between analyzed groups for each drug was calculated as the ratio of median values of LC50 for this drug in each group.

Results of AML patients were published previously [8]. Due to similar profiles of drug sensitivity, all AML patients were pooled into one group for further analysis [8].

Statistical analysis

The Mann-Whitney U test was performed to compare differences in drug resistance between groups.

Results

In comparison to adult AML, CML blasts were more resistant to bortezomib (6.2-fold; p < 0.001), and to the following other drugs: prednisolone (1.5-fold; p = 0.037), vincristine (2.3; p = 0.004), doxorubicin (> 6.9; p < 0.001), etoposide

(7.4; p < 0.001), melphalan (5.9; p = 0.001), cytarabine

(12.5; p = 0.005), fludarabine (2.6; p = 0.008), thiotepa

(5.4; p = 0.001), 4-HOO-cyclophosphamide (2.3; p = 0.015), thioguanine (> 4; p < 0.001), topotecan (20; p < 0.001), and clofarabine (50; p < 0.001). No differences in sensitivity were found for idarubicin, daunorubicin, mitoxantrone, L-asparaginase, cladribine, and treosulfan, while CML cells were 2-fold more sensitive to busulfan (p = 0.035) (Table 1).

CML patients were divided into subgroups (Table 2). No differences in LC50 values for bortezomib were observed between any subgroup of patients. Overall, no significant differences for all tested drugs, including TKIs, were observed between CML patients with non-advanced and advanced disease. CML patients with poor clinical response expressed as clinical resistance to imatinib had higher median LC50 values for vincristine (2.5-fold; p = 0.016), daunorubicin (3.1-fold;

p = 0.011), etoposide (2.2-fold; p = 0.031), and busulfan

(4.5-fold; p = 0.032). No significant differences were observed with respect to other drugs, including all 3 TKIs. CML patients with mutation had higher median LC50 values for vincristine (3.3-fold; p = 0.044), idarubicin (> 7.9-fold; p = 0.031), thiotepa (13.7-fold; p = 0.044), and busulfan (21.6-fold;

p = 0.024). No significant differences were observed with respect to other drugs, including all 3 TKIs (Table 2).

Discussion

Therapy of CML has been significantly improved with the use of BCR-ABL kinase inhibitors. However, the existence of CML cells that are unaffected by BCR-ABL inhibition represents a major barrier that may prevent curative therapy with the current approaches. To date, it seems that resistance to tyrosine kinase inhibitor-based therapies involving BCR-ABL gene mutations and amplification is the most important mechanism of therapy failure. New evidence suggests that persistence of CML stem cells or acquisition of stem cell-like characteristics may prevent complete elimination of CML by TKIs [9]. New targets should be defined before significant progress in curative therapies is possible. The proteasome inhibitor bortezomib is a potent in vitro cytotoxic compound against stem cells in acute and chronic myeloid leukemias [10, 11]. Poor therapy outcome, especially in patients with relapsed and refractory leukemia, might be related to intrinsic drug resistance.

In our previous ex vivo analysis we showed the benefit of use of bortezomib in adult patients with relapsed/refractory AML [8]. Differences in in vitro sensitivity of leukemic cells to bortezomib are related to variability in the activity profiles of the individual proteasomal subunits between primary leukemia cells. In addition to drug resistance, an aberrant activation

of signal transduction proteins, including the NF-B pathway, is one of the key mechanisms of treatment failure in AML

[12, 13]. Activity of bortezomib in AML and CML, which also acts through the NF-B pathway, is an important aspect, being investigated in both in vitro and in vivo studies [14, 15].

BCR-ABL plays an essential role in the pathogenesis of CML and some cases of ALL. Although ABL kinase inhibitors have shown great promise in the treatment of CML, the persistence of residual disease and the occurrence of resistance have prompted investigations into the molecular effectors of BCR-ABL.

Jagani et al. [16] provided a novel insight into the molecular effects of proteasome inhibitor therapy and showed that BCR-ABL stimulated the proteasome-dependent degradation of members of the forkhead family of tumor suppressors in vitro, in an in vivo animal model, and in samples from patients with BCR-ABL-positive CML. They showed that inhibition of this pathway, using bortezomib, caused regression of CML disease. Bortezomib treatment led to inhibition of BCR-ABL-induced suppression of FoxO proteins and their proapoptotic targets, and tumor necrosis factor-related apoptosis-inducing ligand. Their study provided evidence that bortezomib induced apoptosis of CML cells in vitro and might be a candidate therapeutic in the treatment of BCR-ABL-induced leukemia.

Our study, based on the MTT assay, which is an endpoint type analysis, has shown that in comparison to AML cells, bortezomib alone has little ex vivo activity against CML cells. This was observed both for the whole group and for all subsets of patients tested in the study. Recently published results of a pilot study of bortezomib therapy for patients with imatinib-refractory chronic myeloid leukemia in chronic or accelerated phase, performed in the MD Anderson Cancer Center in Houston, have also shown only minimal efficacy, but considerable toxicity in patients with imatinib-refractory CML [14].

The introduction of BCR-ABL1 tyrosine kinase inhibitors during the last decade resulted in long-term disease control in the majority of patients with CML. In those who fail to respond and/or develop intolerance to these agents, still transplantation remains the only effective therapeutic solution [17]. Possibly, combined use of a tyrosine kinase inhibitor and proteasome inhibitor might be helpful for optimizing treatment of refractory/resistant CML [18]. New possibilities can arise with new modalities, related to immunotherapy or other targeted therapy [19, 20]. Further studies should focus on alternative approaches in using proteasome inhibitors in the

treatment of CML, such as in combination with TKIs or as a strategy to eradicate leukemic stem cells [18, 21].

Acknowledgements

This study was supported by the following grants: EC 2008/2009 ZPORR SPS.IV-3040-UE/217/2009; EFS 9/9/POKL /4.4.1/2008; UMK 09/2009 and MNiSW N407 078 32/2964.

References

 1. Bosanquet AG, Nygren P, Weisenthal L. Individualized Tumor Response Testing in Leukemia and Lymphoma. In: Innovative Leukemia and Lymphoma Therapy. Kaspers GJL, Coiffier B, Hein-

rich MC, Estey E (eds.). Informa Healthcare, New York 2008.

 2. Styczynski J, Gil L, Derwich K, et al. Comparison of clofarabine activity in childhood and adult acute leukemia: individual tumor response study. Anticancer Res 2009; 29: 1643-50.

 3. Tipping AJ, Mahon FX, Zafirides G, Lagarde V, Goldman JM, Melo JV. Drug responses of imatinib mesylate-resistant cells: synergism of imatinib with other chemotherapeutic drugs. Leukemia 2002; 16: 2349-57.

 4. Kawaguchi H, Taketani T, Hongo T, et al. In vitro drug resistance to imatinib and mutation of ABL gene in childhood Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia. Leuk Lymphoma 2005; 46: 273-6.

 5. Redaelli S, Piazza R, Rostagno R, Magistroni V, Perini P, Marega M, Gambacorti-Passerini C, Boschelli F. Activity of bosutinib, dasatinib, and nilotinib against 18 imatinib-resistant BCR/ABL mutants. J Clin Oncol 2009; 27: 469-71.

 6. Gil L, Lewandowski K, Komarnicki M. Transplantacja alogenicznych komórek hematopoetycznych w leczeniu przewlekłej białaczki szpikowej – współczesne poglądy w dobie stosowania inhibitorów kinazy tyrozynowej. Wsp Onkol 2010; 14: 31-8.

 7. Baccarani M, Cortes J, Pane F, et al. Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. J Clin Oncol 2009; 27: 6041-51.

 8. Gil L, Styczynski J, Dytfeld D et al. Activity of bortezomib in adult de novo and relapsed acute myeloid leukemia. Anticancer Res 2007; 27: 4021-5.

 9. Donato NJ, Peterson LF. Chronic myeloid leukemia stem cells and developing therapies. Leuk Lymphoma 2011; 52 Suppl 1: 60-80.

10. Servida F, Soligo D, Delia D, Henderson C, Brancolini C, Lombardi L, Deliliers GL. Sensitivity of human multiple myelomas and myeloid leukemias to the proteasome inhibitor I. Leukemia 2005; 19: 2324-31.

11. Yong AS, Keyvanfar K, Hensel N et al. Primitive quiescent CD34+ cells in chronic myeloid leukemia are targeted by in vitro expanded natural killer cells, which are functionally enhanced by bortezomib. Blood 2009; 113: 875-82.

12. Guzman ML, Neering SJ, Upchurch D, Grimes B, Howard DS, Riz-

zieri DA, Luger SM, Jordan CT. Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood 2001; 98: 2301-7.

13. Turco MC, Romano MF, Petrella A, Bisogni R, Tassone P, Venuta S.

NF-kappaB/Rel-mediated regulation of apoptosis in hematologic malignancies and normal hematopoietic progenitors. Leukemia 2004;

18: 11-17.

14. Santos FP, Kantarjian H, McConkey D, et al. Pilot study of bortezomib for patients with imatinib-refractory chronic myeloid leukemia in chronic or accelerated phase. Clin Lymphoma Myeloma Leuk 2011; 11: 355-60.

15. Attar EC, De Angelo DJ, Supko JG, et al. Phase I and pharmacokinetic study of bortezomib in combination with idarubicin and cytarabine in patients with acute myelogenous leukemia. Clin Cancer Res 2008; 14: 1446-1454.

16. Jagani Z, Song K, Kutok JL et al. Proteasome inhibition causes regression of leukemia and abrogates BCR-ABL-induced evasion of apoptosis in part through regulation of forkhead tumor suppressors. Cancer Res 2009; 69: 6546-55.

17. Pavlu J, Szydlo RM, Goldman JM, Apperley JF. Three decades of transplantation for chronic myeloid leukemia: what have we learned? Blood 2011; 117: 755-63.

18. Hu Z, Pan XF, Wu FQ et al. Synergy between proteasome inhibitors and imatinib mesylate in chronic myeloid leukemia. PLoS One 2009; 4: e6257.

19. Mackiewicz J, Mackiewicz A. Immunoterapia nowotworów i perspektywy jej rozwoju. Wspolczesna Onkol 2010; 14: 59-71.

20. Mackiewicz J, Kwinta Ł. Nowe terapie celowane stosowane u chorych na czerniaka uogólnionego. Wspolczesna Onkol 2010; 14: 15-22.

21. Yong AS, Keyvanfar K, Eniafe R, Savani BN, Rezvani K, Sloand EM, Goldman JM, Barrett AJ. Hematopoietic stem cells and progenitors of chronic myeloid leukemia express leukemia-associated antigens: implications for the graft-versus-leukemia effect and peptide vaccine-based immunotherapy. Leukemia 2008; 22: 1721-7.



Adress for correspondence



prof. dr hab. n. med. Jan Styczyński

Katedra i Klinika Pediatrii, Hematologii i Onkologii

Collegium Medicum im. L. Rydygiera w Bydgoszczy

Uniwersytet Mikołaja Kopernika

ul. M. Curie-Skłodowskiej 9

85-094 Bydgoszcz

tel. +48 52 585 4860

fax +48 52 585 4867

e-mail: jstyczynski@cm.umk.pl



Submitted: 11.09.2011

Accepted: 31.01.2012
Copyright: © 2012 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.