1
|
Hattori EY, Masuda T, Mineharu Y, Mikami M, Terada Y, Matsui Y, Kubota H, Matsuo H, Hirata M, Kataoka TR, Nakahata T, Ikeda S, Miyamoto S, Sugiyama H, Arakawa Y, Kamikubo Y. A RUNX-targeted gene switch-off approach modulates the BIRC5/PIF1-p21 pathway and reduces glioblastoma growth in mice. Commun Biol 2022; 5:939. [PMID: 36085167 PMCID: PMC9463152 DOI: 10.1038/s42003-022-03917-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/30/2022] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma is the most common adult brain tumour, representing a high degree of malignancy. Transcription factors such as RUNX1 are believed to be involved in the malignancy of glioblastoma. RUNX1 functions as an oncogene or tumour suppressor gene with diverse target genes. Details of the effects of RUNX1 on the acquisition of malignancy in glioblastoma remain unclear. Here, we show that RUNX1 downregulates p21 by enhancing expressions of BIRC5 and PIF1, conferring anti-apoptotic properties on glioblastoma. A gene switch-off therapy using alkylating agent-conjugated pyrrole-imidazole polyamides, designed to fit the RUNX1 DNA groove, decreased expression levels of BIRC5 and PIF1 and induced apoptosis and cell cycle arrest via p21. The RUNX1-BIRC5/PIF1-p21 pathway appears to reflect refractory characteristics of glioblastoma and thus holds promise as a therapeutic target. RUNX gene switch-off therapy may represent a novel treatment for glioblastoma. Interfering with RUNX family proteins reduces glioblastoma growth in mice and reveals pathways involved in the maintenance of tumour growth.
Collapse
|
2
|
Lee GW, Kang MH, Jeon JH, Song DW, Ro WB, Kim HS, Park HM. Case Report: Long-Term Survival of a Dog With Chronic Lymphocytic Leukemia Treated With Chlorambucil, Prednisolone, and Imatinib. Front Vet Sci 2022; 8:625527. [PMID: 35111836 PMCID: PMC8801678 DOI: 10.3389/fvets.2021.625527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
A 7-year-old castrated male Poodle dog presented with chronic progressive lymphocytosis. Hematologic and peripheral blood smear findings included remarkable lymphocytosis with well-differentiated small lymphocytes. Cytology of bone marrow aspirate showed hypercellular integrity with infiltration of small mature lymphocytes, accounting for 45% of all nucleated cells. Flow cytometry of blood and marrow samples revealed neoplastic lymphocytes predominantly expressing the CD21 molecule. B-cell chronic lymphocytic leukemia (CLL) was diagnosed on an immunophenotypic analysis. Administrations of prednisolone and chlorambucil were initiated and the response was unremarkable. Therefore, additional treatment with imatinib was provided, which resolved the hematologic abnormalities associated with CLL. Flow cytometry after ~1 year of treatment showed normalization of the count of lymphocytes positive for CD21 and resolved hematologic lymphocytosis. The dog was followed-up for 2 years, and there were no severe adverse effects. This case indicates that imatinib may be a good option as an adjunctive therapy with prednisolone and chlorambucil treatment for CLL in dogs without treatment response.
Collapse
Affiliation(s)
- Ga-Won Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Min-Hee Kang
- Department of Bio-Animal Care, Jangan University, Hwaseong, South Korea
| | - Jin-Ha Jeon
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Doo-Won Song
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Woong-Bin Ro
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Heyong-Seok Kim
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Hee-Myung Park
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
- *Correspondence: Hee-Myung Park
| |
Collapse
|
3
|
Brain-invasive meningiomas: molecular mechanisms and potential therapeutic options. Brain Tumor Pathol 2021; 38:156-172. [PMID: 33903981 DOI: 10.1007/s10014-021-00399-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
Meningiomas are the most commonly diagnosed benign intracranial adult tumors. Subsets of meningiomas that present with extensive invasion into surrounding brain areas have high recurrence rates, resulting in difficulties for complete resection, substantially increased mortality of patients, and are therapeutically challenging for neurosurgeons. Exciting new data have provided insights into the understanding of the molecular machinery of invasion. Moreover, clinical trials for several novel approaches have been launched. Here, we will highlight the mechanisms which govern brain invasion and new promising therapeutic approaches for brain-invasive meningiomas, including pharmacological approaches targeting three major aspects of tumor cell invasion: extracellular matrix degradation, cell adhesion, and growth factors, as well as other innovative treatments such as immunotherapy, hormone therapy, Tumor Treating Fields, and biodegradable copolymers (wafers), impregnated chemotherapy. Those ongoing studies can offer more diversified possibilities of potential treatments for brain-invasive meningiomas, and help to increase the survival benefits for patients.
Collapse
|
4
|
Song Y, Park SY, Wu Z, Liu KH, Seo YH. Hybrid inhibitors of DNA and HDACs remarkably enhance cytotoxicity in leukaemia cells. J Enzyme Inhib Med Chem 2021; 35:1069-1079. [PMID: 32314611 PMCID: PMC7191901 DOI: 10.1080/14756366.2020.1754812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Chlorambucil is a nitrogen mustard-based DNA alkylating drug, which is widely used as a front-line treatment of chronic lymphocytic leukaemia (CLL). Despite its widespread application and success for the initial treatment of leukaemia, a majority of patients eventually develop acquired resistance to chlorambucil. In this regard, we have designed and synthesised a novel hybrid molecule, chloram-HDi that simultaneously impairs DNA and HDAC enzymes. Chloram-HDi efficiently inhibits the proliferation of HL-60 and U937 leukaemia cells with GI50 values of 1.24 µM and 1.75 µM, whereas chlorambucil exhibits GI50 values of 21.1 µM and 37.7 µM against HL-60 and U937 leukaemia cells, respectively. The mechanism behind its remarkably enhanced cytotoxicity is that chloram-HDi not only causes a significant DNA damage of leukaemia cells but also downregulates DNA repair protein, Rad52, resulting in the escalation of its DNA-damaging effect. Furthermore, chloram-HDi inhibits HDAC enzymes to induce the acetylation of α-tubulin and histone H3.
Collapse
Affiliation(s)
- Yoojin Song
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Sun You Park
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Zhexue Wu
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Kwang-Hyeon Liu
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Young Ho Seo
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
5
|
Alhmoud JF, Woolley JF, Al Moustafa AE, Malki MI. DNA Damage/Repair Management in Cancers. Cancers (Basel) 2020; 12:E1050. [PMID: 32340362 PMCID: PMC7226105 DOI: 10.3390/cancers12041050] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
DNA damage is well recognized as a critical factor in cancer development and progression. DNA lesions create an abnormal nucleotide or nucleotide fragment, causing a break in one or both chains of the DNA strand. When DNA damage occurs, the possibility of generated mutations increases. Genomic instability is one of the most important factors that lead to cancer development. DNA repair pathways perform the essential role of correcting the DNA lesions that occur from DNA damaging agents or carcinogens, thus maintaining genomic stability. Inefficient DNA repair is a critical driving force behind cancer establishment, progression and evolution. A thorough understanding of DNA repair mechanisms in cancer will allow for better therapeutic intervention. In this review we will discuss the relationship between DNA damage/repair mechanisms and cancer, and how we can target these pathways.
Collapse
Affiliation(s)
- Jehad F. Alhmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - John F. Woolley
- Department of Molecular & Clinical Pharmacology, Liverpool University, Liverpool L69 3GE, UK;
| | | | - Mohammed Imad Malki
- College of Medicine, QU Health, Qatar University, Doha P. O. Box 2713, Qatar;
| |
Collapse
|
6
|
Viswanathan K, Roboz G, Chadburn A, Mathew S. Chronic Myelogenous Leukemia Diagnosed in the Setting of Untreated Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. Int J Surg Pathol 2019; 28:216-224. [PMID: 31544558 DOI: 10.1177/1066896919876704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Chronic myeloid leukemia (CML) is rarely reported to occur in treated chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL). In this article, we report a woman in her 70s, diagnosed with CLL/SLL in 2000, untreated, who subsequently presented 12 years later with de novo CML, BCR-ABL1+. Her IGHV mutated CLL/SLL based on the initial sample in our laboratory showed homozygous and heterozygous 13q14.3 deletions, whereas her CML, at presentation, showed a 46,XX,t(9;22)(q34;q11.2)[7]/46,XX[18] karyotype with a p190 BCR-ABL1 transcript. The tumor burden of each clone varied with treatment, including when treated with dasatinib, used to target both clones. In addition, the cytogenetic abnormalities evolved over time and treatments and included acquisition of an extra chromosome 8 in the CML clone and a novel K1992T ATM missense mutation (47% allele frequency) in the CLL/SLL clone. The patient's last bone marrow biopsy, 5 years after her CML diagnosis and 17 years after the CLL/SLL diagnosis, showed residual CML with extensive involvement by CLL/SLL (80%). Cytogenetic studies showed a 46,XX karyotype, while FISH identified 13q14.3 deletion and the BCR-ABL1 translocation in the CLL/SLL and CML clones, respectively. To date, this is the fourth case of concurrent CML, BCR-ABL1+ arising in untreated CLL/SLL. Here we show dynamic variation in the size of the 2 clonal processes reflecting the variable responsiveness to specific therapies. In addition to the unusual BCR-ABL1+ p190 transcript in the patient's CML, a novel ATM K1992T mutation was identified in the CLL/SLL population.
Collapse
Affiliation(s)
| | - Gail Roboz
- NewYork-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| | - Amy Chadburn
- NewYork-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| | - Susan Mathew
- NewYork-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
7
|
Shea LK, Mikhail FM, Forero-Torres A, Davis RS. Concomitant imatinib and ibrutinib in a patient with chronic myelogenous leukemia and chronic lymphocytic leukemia. Clin Case Rep 2017; 5:899-901. [PMID: 28588835 PMCID: PMC5458012 DOI: 10.1002/ccr3.974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 03/10/2017] [Accepted: 03/23/2017] [Indexed: 11/07/2022] Open
Abstract
The availability of kinase and other small‐molecule inhibitors to treat hematologic malignancies is increasing. Accordingly, novel regimens that employ these therapeutics are rapidly evolving. Herein we report the safe and effective administration of two targeted kinase inhibitors in a patient with concomitant chronic myelogenous leukemia and chronic lymphocytic leukemia.
Collapse
Affiliation(s)
- Lauren K Shea
- Department of Medicine University of Alabama at Birmingham Birmingham Alabama
| | - Fady M Mikhail
- Department of Genetics University of Alabama at Birmingham Birmingham Alabama
| | - Andres Forero-Torres
- Department of Medicine University of Alabama at Birmingham Birmingham Alabama.,Comprehensive Cancer Center University of Alabama at Birmingham Birmingham Alabama
| | - Randall S Davis
- Department of Medicine University of Alabama at Birmingham Birmingham Alabama.,Comprehensive Cancer Center University of Alabama at Birmingham Birmingham Alabama.,Department of Microbiology University of Alabama at Birmingham Birmingham Alabama.,Department of Biochemistry and Molecular Genetics University of Alabama at Birmingham Birmingham Alabama
| |
Collapse
|
8
|
Samuel J, Jayne S, Chen Y, Majid A, Wignall A, Wormull T, Najeeb H, Luo JL, Jones GDD, Macip S, Dyer MJS. Posttranscriptional Upregulation of p53 by Reactive Oxygen Species in Chronic Lymphocytic Leukemia. Cancer Res 2016; 76:6311-6319. [PMID: 27634759 DOI: 10.1158/0008-5472.can-16-0843] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 08/09/2016] [Indexed: 11/16/2022]
Abstract
Chronic lymphocytic leukemia (CLL) cells multiply and become more resistant to immunochemotherapy in "proliferation centers" within tissues, whereas apoptosis occurs in the periphery. Various models recapitulate these microenvironments in vitro, such as stimulation with CD154 and IL4. Using this system, we observed a 30- to 40-fold induction of wild-type p53 protein in 50 distinct human CLL specimens tested, without the induction of either cell-cycle arrest or apoptosis. In contrast, the mRNA levels for p53 did not increase, indicating that its elevation occurred posttranscriptionally. Mechanistic investigations revealed that under the conditions studied, p53 was phosphorylated on residues associated with p53 activation and increased half-life. However, p53 protein induced in this manner could transcriptionally activate only a subset of target genes. The addition of a DNA-damaging agent further upregulated p53 protein levels, which led to apoptosis. p53 induction relied on the increase in intracellular reactive oxygen species observed after CD154 and IL4 stimulation. We propose that chronic oxidative stress is a characteristic of the microenvironment in B-cell "proliferation centers" in CLL that are capable of elevating the basal expression of p53, but to levels below the threshold needed to induce arrest or apoptosis. Our findings suggest that reactivation of the full transcriptional activities of p53 in proliferating CLL cells may offer a possible therapeutic strategy. Cancer Res; 76(21); 6311-9. ©2016 AACR.
Collapse
Affiliation(s)
- Jesvin Samuel
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom.,Mechanisms of Cancer and Ageing Lab, University of Leicester, Leicester, United Kingdom.,Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom.,CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom
| | - Sandrine Jayne
- Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom.,CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom.,Department of Cancer Studies, University of Leicester, Leicester, United Kingdom
| | - Yixiang Chen
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom.,Mechanisms of Cancer and Ageing Lab, University of Leicester, Leicester, United Kingdom.,Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom.,CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom
| | | | - Alice Wignall
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom.,Mechanisms of Cancer and Ageing Lab, University of Leicester, Leicester, United Kingdom.,Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom.,CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom
| | - Timothy Wormull
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom.,Mechanisms of Cancer and Ageing Lab, University of Leicester, Leicester, United Kingdom.,Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom.,CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom
| | - Hishyar Najeeb
- CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom.,Department of Cancer Studies, University of Leicester, Leicester, United Kingdom.,Department of Clinical Biochemistry, College of Medicine, University of Duhok, Kurdistan Regional Government, Iraq
| | - Jin-Li Luo
- CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom
| | - George D D Jones
- CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom.,Department of Cancer Studies, University of Leicester, Leicester, United Kingdom
| | - Salvador Macip
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom. .,Mechanisms of Cancer and Ageing Lab, University of Leicester, Leicester, United Kingdom.,Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom.,CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom
| | - Martin J S Dyer
- Ernest and Helen Scott Haematological Research Institute, University of Leicester, Leicester, United Kingdom.,CRUK Leicester Centre, University of Leicester, Leicester, United Kingdom.,Department of Cancer Studies, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
9
|
Hydroxyurea with or without imatinib in the treatment of recurrent or progressive meningiomas: a randomized phase II trial by Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO). Cancer Chemother Pharmacol 2015; 77:115-20. [PMID: 26659583 DOI: 10.1007/s00280-015-2927-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
PURPOSE Hydroxyurea (HU) is among the most widely used salvage therapies in progressive meningiomas. Platelet-derived growth factor receptors are expressed in virtually all meningiomas. Imatinib sensitizes transformed cells to the cytotoxic effects of chemotherapeutic agents that interfere with DNA metabolism. The combination of HU with imatinib yielded intriguing results in recurrent malignant glioma. The current trial addressed the activity of this association against meningioma. METHODS Patients with recurrent or progressive WHO grade I-III meningioma, without therapeutic indication for surgery, radiotherapy, or stereotactic radiosurgery, aged 18-75 years, ECOG performance status 0-2, and not on enzyme-inducing anti-epileptic drugs were randomized to receive HU 500 mg BID ± imatinib 400 mg QD until progression, unacceptable toxicity, or patient's refusal. The primary endpoint was progression-free survival rate at 9 months (PFS-9). RESULTS Between September 2009 and February 2012, 15 patients were randomized to receive HU + imatinib (N = 7; Arm A) or HU alone (N = 8; Arm B). Afterward the trial was prematurely closed due to slow enrollment rate. PFS-9 (A/B) was 0/75%, and median PFS was 4/19.5 months. Median and 2-year overall survival (A/B) rates were: 6/27.5 months; 28.5/75%, respectively. Main G3-4 toxicities were: G3 neutropenia in 1/0, G4 headache in 1/1, and G3 vomiting in 1/0. CONCLUSION The conduction of a study in recurrent or progressive meningioma remains a challenge. Given the limited number of patients enrolled, no firm conclusions can be drawn about the combination of imatinib and HU. The optimal systemic therapy for meningioma failing surgery and radiation has yet to be identified.
Collapse
|
10
|
Abu-Sanad A, Wang Y, Hasheminasab F, Panasci J, Noë A, Rosca L, Davidson D, Amrein L, Sharif-Askari B, Aloyz R, Panasci L. Simultaneous inhibition of ATR and PARP sensitizes colon cancer cell lines to irinotecan. Front Pharmacol 2015; 6:147. [PMID: 26257651 PMCID: PMC4510998 DOI: 10.3389/fphar.2015.00147] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 07/02/2015] [Indexed: 11/21/2022] Open
Abstract
Enhanced DNA damage repair is one mechanism involved in colon cancer drug resistance. Thus, targeting molecular components of repair pathways with specific small molecule inhibitors may improve the efficacy of chemotherapy. ABT-888 and VE-821, inhibitors of poly-ADP-ribose-polymerase (PARP) and the serine/threonine-kinase Ataxia telangiectasia related (ATR), respectively, were used to treat colon cancer cell lines in combination with the topoisomerase-I inhibitor irinotecan (SN38). Our findings show that each of these DNA repair inhibitors utilized alone at nontoxic single agent concentrations resulted in sensitization to SN38 producing a 1.4–3 fold reduction in the 50% inhibitory concentration (IC50) of SN38 in three colon cancer cell lines. When combined together, nontoxic concentrations of ABT-888 and VE-821 produced a 4.5–27 fold reduction in the IC50 of SN38 with the HCT-116 colon cancer cells demonstrating the highest sensitization as compared to LoVo and HT-29 colon cancer cells. Furthermore, the combination of all three agents was associated with maximal G2 −M arrest and enhanced DNA-damage (γH2AX) in all three colon cancer cell lines. The mechanism of this enhanced sensitization was associated with: (a) maximal suppression of SN38 induced PARP activity in the presence of both inhibitors and (b) ABT-888 producing partial abrogation of the VE-821 enhancement of SN38 induced DNA-PK phosphorylation, resulting in more unrepaired DNA damage; these alterations were only present in the HCT-116 cells which have reduced levels of ATM. This novel combination of DNA repair inhibitors may be useful to enhance the activity of DNA damaging chemotherapies such as irinotecan and help produce sensitization to this drug in colon cancer.
Collapse
Affiliation(s)
- Atlal Abu-Sanad
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Yunzhe Wang
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Fatemeh Hasheminasab
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Justin Panasci
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Alycia Noë
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Lorena Rosca
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - David Davidson
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Lilian Amrein
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Bahram Sharif-Askari
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Raquel Aloyz
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| | - Lawrence Panasci
- Montreal Centre for Experimental Therapeutics in Cancer, Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University Montréal, QC, Canada
| |
Collapse
|
11
|
Abstract
Dysregulation of DNA damage repair and signalling to cell cycle checkpoints, known as the DNA damage response (DDR), is associated with a predisposition to cancer and affects responses to DNA-damaging anticancer therapy. Dysfunction of one DNA repair pathway may be compensated for by the function of another compensatory DDR pathway, which may be increased and contribute to resistance to DNA-damaging chemotherapy and radiotherapy. Therefore, DDR pathways make an ideal target for therapeutic intervention; first, to prevent or reverse therapy resistance; and second, using a synthetic lethal approach to specifically kill cancer cells that are dependent on a compensatory DNA repair pathway for survival in the context of cancer-associated oxidative and replicative stress. These hypotheses are currently being tested in the laboratory and are being translated into clinical studies.
Collapse
Affiliation(s)
- Nicola J Curtin
- Newcastle University, Northern Institute for Cancer Research, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
12
|
Robak T, Robak E. Tyrosine kinase inhibitors as potential drugs for B-cell lymphoid malignancies and autoimmune disorders. Expert Opin Investig Drugs 2012; 21:921-947. [PMID: 22612424 DOI: 10.1517/13543784.2012.685650] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION In the last few years, several tyrosine kinase inhibitors (TKIs) have been synthesized and become available for preclinical studies and clinical trials. This article summarizes recent achievements in the mechanism of action, pharmacological properties, and clinical activity and toxicity, as well as the emerging role of TKIs in lymphoid malignancies, allergic diseases, and autoimmune disorders. AREAS COVERED A literature review was conducted of the MEDLINE database PubMed for articles in English. Publications from 2000 through January 2012 were scrutinized. The search terms used were Bruton's tyrosine kinase (Btk) inhibitors, PCI-32765, GDC-0834, LFM-A13, AVL-101, AVL-292, spleen tyrosine kinase (Syk) inhibitors, R343, R406, R112, R788, fostamatinib, BAY-61-3606, C-61, piceatannol, Lyn, imatinib, nilotinib, bafetinib, dasatinib, GDC-0834, PP2, SU6656 in conjunction with lymphoid malignancy, NHL, CLL, autoimmune disease, allergic disease, asthma, and rheumatoid arthritis. Conference proceedings from the previous 5 years of the American Society of Hematology, European Hematology Association, American Society of Clinical Oncology, and ACR/ARHP Annual Scientific Meetings were searched manually. Additional relevant publications were obtained by reviewing the references from the chosen articles. EXPERT OPINION The use of TKIs, especially inhibitors of Btk, Syk, and Lyn, is a promising new strategy for targeted treatment of B-cell lymphoid malignancies, autoimmune disorders and allergic diseases. However, definitive data from ongoing and future clinical trials will aid in better defining the status of TKIs in the treatment of these disorders.
Collapse
Affiliation(s)
- Tadeusz Robak
- Medical University of Lodz, Department of Hematology, Lodz, Poland.
| | | |
Collapse
|
13
|
Guaragna A, Chiaviello A, Paolella C, D'Alonzo D, Palumbo G, Palumbo G. Synthesis and evaluation of folate-based chlorambucil delivery systems for tumor-targeted chemotherapy. Bioconjug Chem 2011; 23:84-96. [PMID: 22121907 DOI: 10.1021/bc200410d] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of tumor-targeting drug delivery systems, able to selectively transport cytotoxic agents into the tumor site by exploiting subtle morphological and physiological differences between healthy and malignant cells, currently stands as one of the most attractive anticancer strategies used to overcome the selectivity problems of conventional chemotherapy. Owing to frequent overexpression of folate receptors (FRs) on the surface of malignant cells, conjugation of cytotoxic agents to folic acid (FA) via suitable linkers have demonstrated to enhance selective drug delivery to the tumor site. Herein, the chemical synthesis and biological evaluation of two novel folate-conjugates bearing the anticancer agent chlorambucil (CLB) tethered to either an aminoether (4,7,10-trioxa-1,13-tridecanediamine) or a pseudo-β-dipeptide (β-Ala-ED-β-Ala) linker is reported. The two drug delivery systems have been prepared in high overall yields (54% and 34%) through straightforward and versatile synthetic routes. Evaluation of cell specificity was examined using three leukemic cell lines, undifferentiated U937 (not overexpressing FRs, FR(-)), TPA-differentiated U937 (overexpressing FRs, FR(+)), and TK6 (FR(+)) cells. Both conjugates exhibited high specificity only to FR(+) cells (particularly TK6), demonstrating comparable antitumor activity to CLB in its free form. These data confirm the reliability of folate-based drug delivery systems for targeted antitumor therapy; likewise, they lay the foundations for the development of other folate-conjugates with antitumor potential.
Collapse
Affiliation(s)
- Annalisa Guaragna
- Dipartimento di Chimica Organica e Biochimica, Università di Napoli Federico II, via Cinthia 4, Naples, Italy.
| | | | | | | | | | | |
Collapse
|
14
|
Amrein L, Rachid Z, Jean-Claude B, Soulières D, Aloyz R, Panasci L. ZRF4, a combi-molecule with increased efficacy as compared with the individual components in chronic lymphocytic leukemia lymphocytes in vitro. Leukemia 2011; 25:1512-6. [PMID: 21647162 PMCID: PMC3846538 DOI: 10.1038/leu.2011.110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
15
|
Yoon JY, Kumar R, Aloyz R, Johnston JB. Response of concomitant chronic myelogenous leukemia and chronic lymphocytic leukemia to imatinib mesylate. Leuk Res 2011; 35:e179-80. [PMID: 21802141 DOI: 10.1016/j.leukres.2011.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 06/04/2011] [Accepted: 06/06/2011] [Indexed: 11/26/2022]
|
16
|
Amrein PC. The potential for dasatinib in treating chronic lymphocytic leukemia, acute myeloid leukemia, and myeloproliferative neoplasms. Leuk Lymphoma 2011; 52:754-63. [PMID: 21463117 DOI: 10.3109/10428194.2011.555890] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Dasatinib is a kinase inhibitor that inhibits BCR-ABL, Src family kinases, c-Kit, and platelet-derived growth factor receptor kinase. It is licensed for the first- and second-line treatment of chronic myeloid leukemia and second-line treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia on the basis of BCR-ABL inhibition, but the activity of dasatinib against additional molecular targets may enable treatment of other hematologic disorders. Potential targets for dasatinib in chronic lymphocytic leukemia (CLL) include Lyn (a Src family kinase), ABL, and the associated CD40 pathway. Although dasatinib monotherapy has modest clinical activity in CLL, ongoing studies are evaluating combination treatment. In acute myeloid leukemia (AML), FLT3, Lyn, c-Kit, and BCR-ABL are expressed in a subpopulation of patients. To date, clinical responses to dasatinib in patients with unselected AML have been mixed and larger studies are needed, particularly correlating clinical response to molecular markers. Imatinib has been used successfully to treat patients with chronic eosinophilic disorders with the FIP1L1-PDGFRA fusion kinase; limited clinical data indicate that dasatinib could be active in imatinib-resistant disease. Ongoing clinical studies should further define the value of dasatinib in these disorders.
Collapse
Affiliation(s)
- Philip C Amrein
- Hematology-Oncology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
17
|
Economopoulou P, Pappa V, Papageorgiou S, Dervenoulas J, Economopoulos T. Abnormalities of DNA repair mechanisms in common hematological malignancies. Leuk Lymphoma 2011; 52:567-82. [DOI: 10.3109/10428194.2010.551155] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
18
|
Dual inhibition of the homologous recombinational repair and the nonhomologous end-joining repair pathways in chronic lymphocytic leukemia therapy. Leuk Res 2011; 35:1080-6. [PMID: 21281966 DOI: 10.1016/j.leukres.2011.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 01/07/2011] [Accepted: 01/09/2011] [Indexed: 11/21/2022]
Abstract
Resistance to chlorambucil in chronic lymphocytic leukemia (CLL) has been associated with increased DNA repair. Specifically, inhibition of either c-abl, which modulates Rad51 directed homologous recombination or DNA-PK dependent nonhomologous end joining has been shown to sensitize primary CLL lymphocytes to chlorambucil. Here we report that inhibition of c-abl can result in a compensatory increase in DNA-PK and thus inhibition of both c-abl and DNA-PK optimally sensitizes CLL lymphocytes to chlorambucil. In this paper we report a drug-induced compensatory change between two DNA repair pathways with potential therapeutic implications in CLL therapy.
Collapse
|
19
|
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy characterized by clonal expansion of mature B cells that are resistant to apoptosis. This resistance to apoptosis partly results from Mcl-1 expression because high levels of this protein in CLL cells correlate with poor disease prognosis and resistance to chemotherapy. Thus, understanding the mechanism(s) regulating Mcl-1 expression in CLL cells may be useful in the development of new therapies for this incurable disease. In the present study, we show a strong relationship between c-Abl and Mcl-1 expression in CLL cells. We show that treatment of CLL cells with Abl-specific siRNA or with imatinib, to inhibit c-Abl activity, results in the down-regulation of Mcl-1 protein and mRNA. A major regulator of Mcl-1 gene expression is STAT3. Our data show that CLL cells expressing high levels of c-Abl also show elevated levels of phospho-STAT3, and that STAT3 phosphorylation in CLL cells is dependent on c-Abl activity. However, STAT3 phosphorylation by c-Abl requires activation of nuclear factor-κB, secretion of autocrine interleukin-6, and active protein kinase C. Taken together, our data demonstrate the mechanism involved in c-Abl regulation of Mcl-1 expression in CLL cells, and suggest that c-Abl inhibition has therapeutic application in the treatment of this disease.
Collapse
|
20
|
A phase I study of imatinib mesylate in combination with chlorambucil in previously treated chronic lymphocytic leukemia patients. Cancer Chemother Pharmacol 2010; 68:643-51. [PMID: 21120481 DOI: 10.1007/s00280-010-1530-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 11/14/2010] [Indexed: 01/11/2023]
Abstract
PURPOSE The tyrosine kinase inhibitor, imatinib, has the potential to indirectly inhibit DNA repair. This mechanism of action has been shown to mediate sensitization to chlorambucil in chronic lymphocytic leukemia (CLL). To evaluate this effect in vivo, we performed a phase I study of chlorambucil combined with imatinib in relapsed CLL patients. METHODS The three dose levels studied included imatinib at 300, 400, or 600 mg/day. Imatinib was given on days 1-10, and chlorambucil (8 mg/m(2) daily) was given on days 3-7 of a 28-day cycle (up to 6 cycles). RESULTS Eleven patients participated in this study. Low-grade gastrointestinal toxicities were observed in a dose-dependent manner. Forty-five percent of patients responded (two unconfirmed CRs and three PRs). Two responding patients were fludarabine refractory. The in vitro IC(50) of chlorambucil alone or in the presence of 5 μM imatinib in CLL lymphocytes correlated with the decrease in lymphocyte counts on day 15. Imatinib plasma concentrations achieved in patients were in the range of those effective in in vitro sensitization studies. CONCLUSION The combination of chlorambucil and imatinib in patients with previously treated CLL was well tolerated and showed evidence of clinical efficacy. Based on our results, we recommend the 400 mg daily dose of imatinib on days 1-10 with 8 mg/m(3) chlorambucil on days 3-7 every 28 days as the phase II dose. This represents the first clinical trial examining the potential synergy between a tyrosine kinase inhibitor and a conventional alkylating agent for the treatment of CLL.
Collapse
|
21
|
Zauli G, Voltan R, Bosco R, Melloni E, Marmiroli S, Rigolin GM, Cuneo A, Secchiero P. Dasatinib plus Nutlin-3 shows synergistic antileukemic activity in both p53 wild-type and p53 mutated B chronic lymphocytic leukemias by inhibiting the Akt pathway. Clin Cancer Res 2010; 17:762-70. [PMID: 21106726 DOI: 10.1158/1078-0432.ccr-10-2572] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To analyze the effect of the combination of Dasatinib, a multikinase inhibitor, plus Nutlin-3, a nongenotoxic activator of the p53 pathway, in primary B chronic lymphocytic leukemia (B-CLL) patient samples and B leukemic cell line models. EXPERIMENTAL DESIGN The induction of cytotoxicity was evaluated in both primary B-CLL cell samples (n = 20) and in p53(wild-type) (EHEB, JVM-2) and p53(deleted/mutated) (MEC-2, BJAB) B leukemic cell lines. The role of Akt in modulating leukemic cell survival/apoptosis in response to Dasatinib or Dasatinib + Nutlin-3 was documented by functional experiments carried out using specific pharmacological inhibitors and by overexpression of membrane-targeted constitutively active form of Akt. RESULTS The combination of Dasatinib + Nutlin-3 exhibited a synergistic cytotoxicity in the majority (19 out of 20) of B-CLL samples, including patients carrying 17p- (n = 4), and in both p53(wild-type) and p53(deleted/mutated) B leukemic cell lines. At the molecular level, Dasatinib significantly counteracted the Nutlin-3-mediated induction of the p53 transcriptional targets MDM2 and p21 observed in p53(wild-type) leukemic cells. Conversely, Nutlin-3 did not interfere with the ability of Dasatinib to decrease the phosphorylation levels of ERK1/2, p38/MAPK, and Akt in both p53(wild-type) and p53(deleted/mutated) B leukemic cell lines. A critical role of Akt downregulation in mediating the antileukemic activity of Dasatinib and Dasatinib + Nutlin-3 was demonstrated in experiments carried out by specifically modulating the Akt pathway. CONCLUSIONS These findings suggest that Dasatinib + Nutlin-3 might represent an innovative therapeutic combination for both p53(wild-type) and p53(deleted/mutated) B-CLL.
Collapse
Affiliation(s)
- Giorgio Zauli
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Tavolaro S, Chiaretti S, Messina M, Peragine N, Del Giudice I, Marinelli M, Santangelo S, Mauro FR, Guarini A, Foà R. Gene expression profile of protein kinases reveals a distinctive signature in chronic lymphocytic leukemia and in vitro experiments support a role of second generation protein kinase inhibitors. Leuk Res 2010; 34:733-41. [DOI: 10.1016/j.leukres.2009.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 11/03/2009] [Accepted: 11/05/2009] [Indexed: 12/30/2022]
|
23
|
Reardon DA, Dresemann G, Taillibert S, Campone M, van den Bent M, Clement P, Blomquist E, Gordower L, Schultz H, Raizer J, Hau P, Easaw J, Gil M, Tonn J, Gijtenbeek A, Schlegel U, Bergstrom P, Green S, Weir A, Nikolova Z. Multicentre phase II studies evaluating imatinib plus hydroxyurea in patients with progressive glioblastoma. Br J Cancer 2009; 101:1995-2004. [PMID: 19904263 PMCID: PMC2795431 DOI: 10.1038/sj.bjc.6605411] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background: We evaluated the efficacy of imatinib mesylate in addition to hydroxyurea in patients with recurrent glioblastoma (GBM) who were either on or not on enzyme-inducing anti-epileptic drugs (EIAEDs). Methods: A total of 231 patients with GBM at first recurrence from 21 institutions in 10 countries were enrolled. All patients received 500 mg of hydroxyurea twice a day. Imatinib was administered at 600 mg per day for patients not on EIAEDs and at 500 mg twice a day if on EIAEDs. The primary end point was radiographic response rate and secondary end points were safety, progression-free survival at 6 months (PFS-6), and overall survival (OS). Results: The radiographic response rate after centralised review was 3.4%. Progression-free survival at 6 months and median OS were 10.6% and 26.0 weeks, respectively. Outcome did not appear to differ based on EIAED status. The most common grade 3 or greater adverse events were fatigue (7%), neutropaenia (7%), and thrombocytopaenia (7%). Conclusions: Imatinib in addition to hydroxyurea was well tolerated among patients with recurrent GBM but did not show clinically meaningful anti-tumour activity.
Collapse
Affiliation(s)
- D A Reardon
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Box 3624, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Targeting the WNT/beta-catenin/TCF/LEF1 axis in solid and haematological cancers: Multiplicity of therapeutic options. Eur J Cancer 2009; 45:2759-67. [PMID: 19729298 DOI: 10.1016/j.ejca.2009.08.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 07/30/2009] [Accepted: 08/04/2009] [Indexed: 11/21/2022]
Abstract
Among aberrantly regulated signalling pathways in cancer the WNT/beta-catenin pathway plays an outstanding role, since it was shown to be critically involved in a wide range of neoplasias. While the underlying mechanisms vary, overexpression of WNTs was found to mediate active signalling in some of these diseases. Other cancers show a mutation in pathway members further downstream, such as APC, Axin or beta-catenin, leading to aberrant signalling activation. Another mechanism initiating activation of WNT/beta-catenin signalling is the silencing of expression of negative WNT/beta-catenin regulators, such as DKK and WIF1, by, for example, promoter hypermethylation. All these mechanisms result in a common consequence, the activation of TCF/LEF1 transcription factors and subsequent target gene expression. Several target genes are known to be key players in tumourigenesis, such as c-myc, cyclin D1 or survivin. The variety of possible underlying mechanisms leading to beta-catenin/TCF/LEF1 activation offers multiple options to target the aberrantly activated pathway in order to prevent target gene expression and/or their gene products to exert their tumourigenic function. Here, we summarise the physiological role of WNT/beta-catenin signalling and the consequences of its aberrant activation during tumourigenesis. Furthermore, we discuss the possible strategies to target this pathway and their potential importance in cancer treatment.
Collapse
|
25
|
Amrein L, Panasci L, Gibson SB, Johnston JB, Soulières D, Aloyz R. Primary del 17 chronic lymphocytic leukaemia lymphocytes are hypersensitive to dasatinib in vitro. Br J Haematol 2009; 147:396-8. [PMID: 19758122 PMCID: PMC2774145 DOI: 10.1111/j.1365-2141.2009.07814.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Lilian Amrein
- Lady Davis Institute for Medical Research-Cancer Segal Center, Sir M.B. Davis-Jewish General HospitalMontreal, QC
- Montreal Center for Experimental Therapeutics, McGill UniversityMontreal, QC
| | - Lawrence Panasci
- Lady Davis Institute for Medical Research-Cancer Segal Center, Sir M.B. Davis-Jewish General HospitalMontreal, QC
- Montreal Center for Experimental Therapeutics, McGill UniversityMontreal, QC
| | - Spencer B Gibson
- Manitoba Institute of Cell Biology, Cancer Care ManitobaWinnipeg, MB
| | - James B Johnston
- Manitoba Institute of Cell Biology, Cancer Care ManitobaWinnipeg, MB
| | - Denis Soulières
- Department of Medicine, Service of Hematology and Medical Oncology, Centre Hospitalier de l’Université de Montréal and Centre de Recherche du CHUMSherbrooke East, Montreal, QC, Canada E-mail:
| | - Raquel Aloyz
- Lady Davis Institute for Medical Research-Cancer Segal Center, Sir M.B. Davis-Jewish General HospitalMontreal, QC
- Montreal Center for Experimental Therapeutics, McGill UniversityMontreal, QC
| |
Collapse
|
26
|
Abstract
The mainstay of therapy of chronic lymphocytic leukemia (CLL) is cytotoxic chemotherapy; however, CLL is still an incurable disease with resistance to therapy developing in the majority of patients. In recent years, our understanding of the biological basis of CLL pathogenesis has substantially improved and novel treatment strategies are emerging. Tailoring and individualizing therapy according to the molecular and cellular biology of the disease is on the horizon, and advances with targeted agents such as monoclonal antibodies combined with traditional chemotherapy have lead to improved remission rates. The proposed key role of the B-cell receptor (BCR) in CLL pathogenesis has led to a number of possible opportunities for therapeutic exploitation. We are beginning to understand that the microenvironment is of utmost importance in CLL because certain T-cell subsets and stromal cells support the outgrowth and development of the malignant clone. Furthermore, an increase in our understanding of the deregulated cell-death machinery in CLL is a prerequisite to developing new targeted strategies that might be more effective in engaging with the cell-death machinery. This Review summarizes the progress made in understanding these features of CLL biology and describes novel treatment strategies that have also been exploited in current clinical trials.
Collapse
|
27
|
Choudhury A, Zhao H, Jalali F, Al Rashid S, Ran J, Supiot S, Kiltie AE, Bristow RG. Targeting homologous recombination using imatinib results in enhanced tumor cell chemosensitivity and radiosensitivity. Mol Cancer Ther 2009; 8:203-13. [PMID: 19139130 DOI: 10.1158/1535-7163.mct-08-0959] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RAD51 is a key protein in the homologous recombination (HR) pathway of DNA double-strand break repair, and HR represents a novel target for cancer therapy. Because imatinib (Gleevec) has been reported to reduce RAD51 protein levels, we tested the clonogenic survival for RT112, H1299, PANC1, and PC3 tumor cell lines of varying p53 status and normal GM05757 normal fibroblasts after exposure to single agent imatinib (0-20 micromol/L; 0-72 hours). We also combined imatinib with DNA damaging agents that are toxic to RAD51-deficient cells, including ionizing radiation, gemcitabine, and mitomycin C. We observed decreased nuclear expression and chromatin binding of RAD51 protein following imatinib treatment. Imatinib also resulted in decreased error-free HR as determined by a flow cytometry-based integrated direct repeat-green fusion protein reporter system; this correlated to reduced RAD51 expression. Clonogenic survival experiments revealed increased cell kill for imatinib-treated cells in combination with ionizing radiation, gemcitabine, and mitomycin C, due in part to mitotic catastrophe. In experiments using imatinib and gemcitabine, tumor cell lines were sensitized to a greater extent than normal fibroblasts. This preservation of the therapeutic ratio was confirmed in vivo using PC3 xenograft growth delay and intestinal crypt cell clonogenic assays. HR inhibition may be an additional mechanism of action for the chemosensitization and radiosensitization of solid tumors with imatinib with preservation of the therapeutic ratio.
Collapse
Affiliation(s)
- Ananya Choudhury
- Department of Medical Biophysics, University of Toronto and Radiation Medicine Program, Princess Margaret Hospital, 610 University Avenue, Toronto, Ontario, Canada M5G2M9
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Amrein L, Hernandez TA, Ferrario C, Johnston J, Gibson SB, Panasci L, Aloyz R. Dasatinib sensitizes primary chronic lymphocytic leukaemia lymphocytes to chlorambucil and fludarabine in vitro. Br J Haematol 2009; 143:698-706. [PMID: 19062342 DOI: 10.1111/j.1365-2141.2008.07418.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The dual c-abl/Src kinase inhibitor, dasatinib, utilized to treat chronic myeloid leukaemia (CML) when used at clinically attainable sublethal concentrations, synergistically sensitized primary chronic lymphocytic leukaemia (CLL) lymphocytes to chlorambucil and fludarabine. In contrast, dasatinib alone demonstrated toxicity to CLL lymphocytes at concentrations that are generally not clinically attainable. Dasatinib resistance and poorer dasatinib-mediated sensitization to chlorambucil and fludarabine was associated with higher expression of c-abl protein levels. In contrast, chlorambucil and fludarabine resistance correlated with basal p53 protein levels. Moreover, Western blot analysis after in vitro treatment of primary CLL lymphocytes with dasatinib, chlorambucil and/or fludarabine, showed that dasatinib: (i) inhibited c-abl function (e.g. downregulation of c-abl protein levels and decreased the phosphorylation of a c-abl downstream target, Dok2), (ii) decreased chlorambucil/fludarabine induced accumulation of p53 protein levels, (iii) altered the response to chlorambucil/fludarabine induced DNA-damage as evidenced by an increase in chlorambucil/fludarabine-induced H2AX phosphorylation, and (iv) accentuated the c-abl downregulation induced by chlorambucil/fludarabine. Our results suggest that dasatinib in combination with chlorambucil or fludarabine may improve the therapy of CLL.
Collapse
Affiliation(s)
- Lilian Amrein
- Lady Davis Institute for Medical Research-Cancer Segal Center, Sir MB Davis-Jewish General Hospital, Montreal, Canada
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Chronic lymphocytic leukaemia (CLL) is the most common adult leukaemia in Europe and North America. The disease is characterized by proliferation and accumulation of small CD5+ B cells in blood, lymph nodes, spleen, liver and bone marrow. The natural clinical course of CLL is highly variable, and chemotherapy is usually not indicated in early and stable disease. However, patients with progressive and more advanced CLL require treatment. For many years, chlorambucil with or without corticosteroids was used in previously untreated patients with CLL. More recently, purine nucleoside analogues (PNAs) [fludarabine, cladribine and pentostatin] have been included in treatment approaches for this disease, and chlorambucil is no longer the leading standard everywhere. Currently, this drug is rather recommended for the treatment of older, unfit patients with co-morbidities, especially in European countries. Significantly higher overall response (OR) and complete response (CR) rates in patients treated initially with PNAs than in those treated with chlorambucil or cyclophosphamide-based combination regimens have been confirmed in randomized, prospective, multicentre trials. Moreover, PNAs administered in combination with cyclophosphamide produce higher response rates, including CR and molecular CR, compared with PNA as monotherapy. Recent reports suggest that the administration of monoclonal antibodies (mAbs) can significantly improve the course of CLL. At present, two mAbs have the most important clinical value in patients with CLL. The first is rituximab, a human mouse antibody that targets CD20 antigens, and the second is alemtuzumab, a humanized form of a rat antibody active against CD52. Several recent reports suggest that in patients with CLL, rituximab combined with a PNA can increase the OR and CR rates compared with PNA or rituximab alone, with acceptable toxicity. In randomized trials, the combination of rituximab with fludarabine and cyclophosphamide (FC-R regimen) demonstrated higher rates of OR, CR and progression-free survival in patients with previously untreated and relapsed or refractory CLL than fludarabine plus cyclophosphamide (FC regimen). Several reports have confirmed significant activity with alemtuzumab in relapsed or refractory CLL, as well as in previously untreated patients. Recently, several new agents have been investigated and have shown promise in treating patients with CLL. These treatments include new mAbs, agents targeting the antiapoptotic bcl-2 family of proteins and receptors involved in mediating survival signals from the microenvironment, antisense oligonucleotides and other agents. The most promising are new mAbs directed against the CD20 molecule, lumiliximab and anti-CD40 mAbs. Oblimersen, alvocidib (flavopiridol) and lenalidomide are also being evaluated both in preclinical studies and in early clinical trials. In recent years, a significant improvement in haematopoietic stem cell transplantation (HSCT) procedures in patients with high-risk CLL has been observed. However, the exact role of HSCT, autologous or allogeneic, in the standard management of CLL patients is still undefined.
Collapse
MESH Headings
- Adult
- Alemtuzumab
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/therapeutic use
- Antineoplastic Agents/immunology
- Antineoplastic Agents/therapeutic use
- Disease Models, Animal
- Disease-Free Survival
- Europe
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell
- Male
- Mice
- North America
- Rats
Collapse
Affiliation(s)
- Tadeusz Robak
- Department of Hematology, Medical University of Lode, Copernicus Memorial Hospital, Lodz, Poland.
| | | | | |
Collapse
|
30
|
The kinase inhibitor dasatinib induces apoptosis in chronic lymphocytic leukemia cells in vitro with preference for a subgroup of patients with unmutated IgVH genes. Blood 2008; 112:1443-52. [PMID: 18550857 DOI: 10.1182/blood-2007-11-123984] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Src family kinases (SFKs) were described to be overexpressed in chronic lymphocytic leukemia (CLL). We wished to examine the effects of the Src and Abl kinase inhibitor dasatinib on the intracellular signaling and survival of CLL cells. Dasa-tinib showed a dose- and time-dependent reduction of global tyrosine phosphorylation and of activating phosphotyrosine levels of SFKs. Treatment with 100 nM dasatinib led to decreased levels of the activated, phosphorylated forms of Akt, Erk1/2, and p38, and induced PARP cleavage through caspase activity. In Mec1 and JVM-3 cell lines, dasatinib increased p53 protein levels and inhibited proliferation. In freshly isolated CLL cells, dasatinib reduced the expression of Mcl-1 and Bcl-x(L). Combination of 5 microM dasatinib and fludarabine increased the apoptosis induction of each by approximately 50%. In 15 primary CLL samples, cells with unmutated immunoglobulin variable heavy chain (IgV(H)) genes were more sensitive to dasatinib than those with mutated IgV(H) genes (P = .002). In summary, dasatinib shows potent inhibitory effects on the survival of CLL cells in vitro, most prominently in samples obtained from patients with unfavorable prognostic features.
Collapse
|
31
|
Reardon DA, Desjardins A, Vredenburgh JJ, Sathornsumetee S, Rich JN, Quinn JA, Lagattuta TF, Egorin MJ, Gururangan S, McLendon R, Herndon JE, Friedman AH, Salvado AJ, Friedman HS. Safety and pharmacokinetics of dose-intensive imatinib mesylate plus temozolomide: phase 1 trial in adults with malignant glioma. Neuro Oncol 2008; 10:330-40. [PMID: 18359865 DOI: 10.1215/15228517-2008-003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We determined the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT) of imatinib mesylate, an inhibitor of the receptor tyrosine kinases platelet-derived growth factor receptor (PDGFR), the proto-oncogene product c-kit, and the fusion protein Bcr-Abl, when administered for 8 days in combination with temozolomide (TMZ) to malignant glioma (MG) patients. MG patients who had not failed prior TMZ were eligible to receive TMZ at a dose of 150-200 mg/m(2) per day on days 4-8 plus imatinib mesylate administered orally on days 1-8 of each 4-week cycle. Patients were stratified based on concurrent administration of CYP3A4-inducing antiepileptic drugs (EIAEDs). The imatinib dose was escalated in successive cohorts of patients independently for each stratum. Imatinib, at doses ranging from 400 mg to 1,200 mg, was administered with TMZ to 65 patients: 52 (80%) with glioblastoma multiforme (GBM) and 13 (20%) with grade III MG. At enrollment, 34 patients (52%) had stable disease, and 33 (48%) had progressive disease; 30 patients (46%) were on EIAEDs. The MTD of imatinib for patients concurrently receiving or not receiving EIAEDs was 1,000 mg. DLTs were hematologic, gastrointestinal, renal, and hepatic. Pharmacokinetic analyses revealed lowered exposures and enhanced clearance among patients on EIAEDs. Among GBM patients with stable disease at enrollment (n=28), the median progression-free and overall survival times were 41.7 and 56.1 weeks, respectively. Imatinib doses up to 1,000 mg/day for 8 consecutive days are well tolerated when combined with standard TMZ dosing for MG patients. A subsequent phase 2 study is required to further evaluate the efficacy of this regimen for this patient population.
Collapse
Affiliation(s)
- David A Reardon
- Department of Surgery, Duke University Medical Center, Box 3624, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Miyagawa K. Clinical relevance of the homologous recombination machinery in cancer therapy. Cancer Sci 2008; 99:187-94. [PMID: 17953711 PMCID: PMC11160033 DOI: 10.1111/j.1349-7006.2007.00644.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 09/07/2007] [Accepted: 09/09/2007] [Indexed: 12/12/2022] Open
Abstract
Cancer chemotherapy and radiotherapy kill cancer cells by inducing DNA damage, unless the lesions are repaired by intrinsic repair pathways. DNA double-strand breaks (DSB) are the most deleterious type of damage caused by cancer therapy. Homologous recombination (HR) is one of the major repair pathways for DSB and is thus a potential target of cancer therapy. Cells with a defect in HR have been shown to be sensitive to a variety of DNA-damaging agents, particularly interstrand crosslink (ICL)-inducing agents such as mitomycin C and cisplatin. These findings have recently been applied to clinical studies of cancer therapy. ERCC1, a structure-specific endonuclease involved in nucleotide excision repair (NER) and HR, confers resistance to cisplatin. Patients with ERCC1-negative non-small-cell lung cancer were shown to benefit from adjuvant cisplatin-based chemotherapy. Imatinib, an inhibitor of the c-Abl kinase, has been investigated as a sensitizer in DNA-damaging therapy, because c-Abl activates Rad51, which plays a key role in HR. Furthermore, proteins involved in HR have been shown to repair DNA damage induced by a variety of other chemotherapeutic agents, including camptothecin and gemcitabine. These findings highlight the importance of HR machinery in cancer therapy.
Collapse
Affiliation(s)
- Kiyoshi Miyagawa
- Department of Radiation Biology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
33
|
Sampath D, Plunkett W. The role of DNA repair in chronic lymphocytic leukemia pathogenesis and chemotherapy resistance. Curr Oncol Rep 2007; 9:361-7. [PMID: 17706164 DOI: 10.1007/s11912-007-0048-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Front-line therapy for chronic lymphocytic leukemia (CLL) with alkylating agents is associated with low rates of complete remission and no improvement in overall survival. The ability of CLL cells to efficiently repair alkylator-induced damage to DNA might explain this lack of response. Novel strategies that inhibit DNA repair, such as combinations of alkylating agents, purine nucleoside analogues, and immunotherapy, have produced durable clinical and molecular remission in both untreated and relapsed CLL. This review evaluates the contribution of DNA repair processes in the development of resistance to chemotherapy and the impact of therapies that exploit the DNA repair capacity of CLL cells to therapeutic advantage.
Collapse
Affiliation(s)
- Deepa Sampath
- Department of Experimental Therapeutics, Unit 71, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | | |
Collapse
|
34
|
Nowak D, Boehrer S, Hochmuth S, Trepohl B, Hofmann W, Hoelzer D, Hofmann WK, Mitrou PS, Ruthardt M, Chow KU. Src kinase inhibitors induce apoptosis and mediate cell cycle arrest in lymphoma cells. Anticancer Drugs 2007; 18:981-95. [PMID: 17704648 DOI: 10.1097/cad.0b013e3281721ff6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Src kinases are involved in multiple cellular contexts such as proliferation, adhesion, tumor invasiveness, angiogenesis, cell cycle control and apoptosis. We here demonstrate that three newly developed dual selective Src/Abl kinase inhibitors (SrcK-I) (AZM559756, AZD0530 and AZD0424) are able to induce apoptosis and cell cycle arrest in BCR-ABL, c-KIT and platelet-derived growth factor-negative lymphoma cell lines. Treatment of DOHH-2, WSU-NHL, Raji, Karpas-299, HUT78 and Jurkat cells with SrcK-I revealed that the tested substances were effective on these parameters in the cell lines DOHH-2 and WSU-NHL, whereas the other tested cell lines remained unaffected. Phosphorylation of Lyn and in particular Lck were affected most heavily by treatment with the SrcK-I. Extrinsic as well as intrinsic apoptosis pathways were activated and elicited unique expressional patterns of apoptosis-relevant proteins such as downregulation of survivin, Bcl-XL and c-FLIP. Protein levels of c-abl were downregulated and Akt phosphorylation was decreased by treatment with SrcK-I. Basal expression levels of c-Myc were notably lower in sensitive cell lines as compared with nonsensitive cell lines, possibly providing an explanation for sensitivity versus resistance against these novel substances. This study provides the first basis for establishing novel SrcK-I as weapons in the arsenal against lymphoma cells.
Collapse
Affiliation(s)
- Daniel Nowak
- Department of Internal Medicine II, Hematology and Oncology, University Hospital, Theodor-Stern-Kai Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Amrein L, Loignon M, Goulet AC, Dunn M, Jean-Claude B, Aloyz R, Panasci L. Chlorambucil cytotoxicity in malignant B lymphocytes is synergistically increased by 2-(morpholin-4-yl)-benzo[h]chomen-4-one (NU7026)-mediated inhibition of DNA double-strand break repair via inhibition of DNA-dependent protein kinase. J Pharmacol Exp Ther 2007; 321:848-55. [PMID: 17351105 DOI: 10.1124/jpet.106.118356] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chlorambucil (CLB) treatment is used in chronic lymphocytic leukemia (CLL) but resistance to CLB develops in association with accelerated repair of CLB-induced DNA damage. Phosphorylated histone H2AX (gammaH2AX) is located at DNA double-strand break (DSB) sites; furthermore, it recruits and retains damage-responsive proteins. This damage can be repaired by nonhomologous DNA end-joining (NHEJ) and/or homologous recombinational repair (HR) pathways. A key component of NHEJ is the DNA-dependent protein kinase (DNA-PK) complex. Increased DNA-PK activity is associated with resistance to CLB in CLL. We used the specific DNA-PK inhibitor 2-(morpholin-4-yl)-benzo[h]chomen-4-one (NU7026) to sensitize CLL cells to chlorambucil. Our results indicate that in a CLL cell line (I83) and in primary CLL-lymphocytes, chlorambucil plus NU7026 has synergistic cytotoxic activity at nontoxic doses of NU7026. CLB treatment results in G(2)/M phase arrest, and NU7026 increases this CLB-induced G(2)/M arrest. Moreover, a kinetic time course demonstrates that CLB-induced DNA-PK activity was inhibited by NU7026, providing direct evidence of the ability of NU7026 to inhibit DNA-PK function. DSBs, visualized as gammaH2AX, were enhanced 24 to 48 h after CLB and further increased by CLB plus NU7026, suggesting that the synergy of the combination is mediated by NU7026 inhibition of DNA-PK with subsequent inhibition of DSB repair.
Collapse
Affiliation(s)
- Lilian Amrein
- Montreal Centre for Experimental Therapeutics in Cancer-Lady Davis Institute for Medical Research, Sir Mortimer B Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
36
|
Lin K, Glenn MA, Harris RJ, Duckworth AD, Dennett S, Cawley JC, Zuzel M, Slupsky JR. c-Abl Expression in Chronic Lymphocytic Leukemia Cells: Clinical and Therapeutic Implications. Cancer Res 2006; 66:7801-9. [PMID: 16885384 DOI: 10.1158/0008-5472.can-05-3901] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
c-Abl is important for normal B-cell development, but little is known about the function of this nonreceptor tyrosine kinase in chronic lymphocytic leukemia (CLL). Therefore, the aim of the present study was to examine the clinical, therapeutic, and pathogenetic importance of c-Abl in this disease. We show that the malignant cells of CLL predominantly express the type 1b splice variant of c-Abl and that the expression of c-Abl protein is higher in CLL cells than in normal peripheral blood B cells. Moreover, we show that the levels of c-Abl protein expression correlate positively with tumor burden and disease stage, and negatively with IgVH mutation. We also show that STI-571, an inhibitor of c-Abl kinase activity, induces apoptosis of CLL cells with high c-Abl expression levels through a mechanism involving inhibition of nuclear factor κB. We conclude that overexpression of c-Abl is likely to play a pathogenetic role in CLL and that STI-571 may be of potential use in the treatment of this disease. (Cancer Res 2006; 66(15): 7801-9)
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/physiology
- B-Lymphocytes/metabolism
- Benzamides
- Genes, Immunoglobulin Heavy Chain
- Humans
- Imatinib Mesylate
- Immunoglobulin Variable Region/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/enzymology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Mutation
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/metabolism
- Piperazines/pharmacology
- Protein Isoforms
- Proto-Oncogene Proteins c-abl/biosynthesis
- Proto-Oncogene Proteins c-abl/genetics
- Pyrimidines/pharmacology
- ZAP-70 Protein-Tyrosine Kinase/biosynthesis
Collapse
Affiliation(s)
- Ke Lin
- Department of Haematology, University of Liverpool, Daulby Street, Liverpool L69 3GA, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Dresemann G. Imatinib and hydroxyurea in pretreated progressive glioblastoma multiforme: a patient series. Ann Oncol 2005; 16:1702-8. [PMID: 16033874 DOI: 10.1093/annonc/mdi317] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Grade IV malignancies of the brain, such as glioblastoma multiforme (GBM), are associated with a dismal prognosis. Autocrine and paracrine loops of platelet-derived growth factor (PDGF) signaling, as well as other signal transduction pathways, have been postulated to play a role in glioblastoma transformation, and molecules involved in these pathways can potentially serve as targets for therapeutic inhibitory agents. Imatinib, an inhibitor of PDGF receptors alpha and beta, as well as other selected tyrosine kinases, is indicated for treatment of chronic myelogenous leukemia (CML) and gastrointestinal stromal tumor (GIST). Unfortunately, imatinib, as with many conventional chemotherapeutic agents, has limited efficacy as monotherapy in GBM. In preclinical studies, the chemotherapeutic agent hydroxyurea is demonstrated to have cytotoxic effects additive with imatinib. PATIENTS AND METHODS We tested the combination of hydroxyurea and imatinib in 30 grade IV progressive GBM patients refractory to chemo- and radiotherapy. All 30 patients were evaluable after a median 19 weeks observation time. RESULTS Combination therapy with imatinib and hydroxyurea resulted in a 20% response rate, including complete and partial responses. Patients experiencing response or stable disease yielded a combined clinical benefit rate of 57%. Median time to progression was 10 weeks and median overall survival was 19 weeks. Three patients continue to survive on combination therapy, with the shortest duration being 106 weeks. Six-month and 2-year progression-free survival rates were 32% and 16%, respectively. CONCLUSION The efficacy results, combined with findings that imatinib and hydroxyurea were well tolerated, suggest that this combination shows promise as therapy for GBM.
Collapse
Affiliation(s)
- G Dresemann
- Franz-Hospital, Onkologische Abteilung, Dülmen, Germany.
| |
Collapse
|
38
|
Xu ZY, Loignon M, Han FY, Panasci L, Aloyz R. Xrcc3 induces cisplatin resistance by stimulation of Rad51-related recombinational repair, S-phase checkpoint activation, and reduced apoptosis. J Pharmacol Exp Ther 2005; 314:495-505. [PMID: 15843498 DOI: 10.1124/jpet.105.084053] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Eukaryotic cells respond to DNA damage by activation of DNA repair, cell cycle arrest, and apoptosis. Several reports suggest that such responses may be coordinated by communication between damage repair proteins and proteins signaling other cellular responses. The Rad51-guided homologous recombination repair system plays an important role in the recognition and repair of DNA interstrand crosslinks (ICLs), and cells deficient in this repair pathway become hypersensitive to ICL-inducing agents such as cisplatin and melphalan. We investigated the possible role of the Rad51-paralog protein Xrcc3 in drug resistance. Xrcc3 overexpression in MCF-7 cells resulted in 1) a 2- to 6-fold resistance to cisplatin/melphalan, 2) a 2-fold increase in drug-induced Rad51 foci, 3) an increased cisplatin-induced S-phase arrest, 4) decreased cisplatin-induced apoptosis, and 5) increased cisplatin-induced DNA synthesis arrest. Interestingly, Xrcc3 overexpression did not alter the doubling time or cell cycle progression in the absence of DNA damage. Furthermore, Xrcc3 overexpression is associated with increased Rad51C protein levels consistent with the known interaction of these two proteins. Our results demonstrate that Xrcc3 is an important factor in DNA cross-linking drug resistance in human tumor cells and suggest that the response of the homologous recombinational repair machinery and cell cycle checkpoints to DNA cross-linking agents is intertwined.
Collapse
Affiliation(s)
- Zhi-Yuan Xu
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, 3755 Côte Ste. Catherine Road, Montreal, QC, Canada H3T 1E2
| | | | | | | | | |
Collapse
|
39
|
Reply to Aloyz et al. Leukemia 2005. [DOI: 10.1038/sj.leu.2403742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Aloyz R, Panasci L. Reply to ‘Imatinib induces apoptosis in CLL lymphocytes with high expression of Par-4’ by Chow et al. Leukemia 2005. [DOI: 10.1038/sj.leu.2403740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
41
|
Katsoulas A, Rachid Z, Brahimi F, McNamee J, Jean-Claude BJ. Cytokinetics and mechanism of action of AKO4: a novel nitrogen mustard targeted to bcr-abl. Leuk Res 2005; 29:565-72. [PMID: 15755509 DOI: 10.1016/j.leukres.2004.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Accepted: 11/09/2004] [Indexed: 10/25/2022]
Abstract
The "combi-targeting" concept seeks to design molecules to not only block tyrosine kinase (TK) activity but also to induce DNA damage. Here we design AK04, a molecule that combines the pharmacophore chlorambucil with that of STI-571 (Gleevec). The results showed that although a less potent abl TK inhibitor than STI571, AK04 was capable of significantly blocking bcr-abl phosphorylation not only in a purified abl assay but also in the bcr-abl+ K562 cells. In contrast to STI571 and like chlorambucil, it induced a dose-dependent increase in DNA damage in these cells. More importantly, AK04 was 12-32-fold more potent than chlorambucil in all bcr-abl+ cells of our cell panel. In the isogenic human megakaryocytic Mo7e and Mo7/bcr-abl cells, AK04 selectively killed the bcr-abl transfectants. Flow cytometry revealed that despite being a five-fold less potent inhibitor of bcr-abl than STI-571, it induced a significant dose-dependent increase in levels of cell death by apoptosis in KU812 cells 24 h post-treatment. Under these conditions, chlorambucil did not induce any significant level of apoptosis. These results suggest that AK04 is a nitrogen mustard with binary bcr-abl/DNA targeting effects, a property that may account for its superior potency when compared with the classical mustard chlorambucil.
Collapse
Affiliation(s)
- Athanasia Katsoulas
- Cancer Drug Research Laboratory, Department of Medicine, Division of Medical Oncology, McGill University Health Center/Royal Victoria Hospital, 687 Pine Ave. West, M7.19, Montreal, Que., Canada H3A 1A1
| | | | | | | | | |
Collapse
|
42
|
Chow KU, Nowak D, Hofmann W, Schneider B, Hofmann WK. Imatinib induces apoptosis in CLL lymphocytes with high expression of Par-4. Leukemia 2005; 19:1103-5; author reply 1105-6; discussion 1106-7. [PMID: 15815724 DOI: 10.1038/sj.leu.2403739] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Drabløs F, Feyzi E, Aas PA, Vaagbø CB, Kavli B, Bratlie MS, Peña-Diaz J, Otterlei M, Slupphaug G, Krokan HE. Alkylation damage in DNA and RNA--repair mechanisms and medical significance. DNA Repair (Amst) 2005; 3:1389-407. [PMID: 15380096 DOI: 10.1016/j.dnarep.2004.05.004] [Citation(s) in RCA: 462] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Indexed: 12/13/2022]
Abstract
Alkylation lesions in DNA and RNA result from endogenous compounds, environmental agents and alkylating drugs. Simple methylating agents, e.g. methylnitrosourea, tobacco-specific nitrosamines and drugs like temozolomide or streptozotocin, form adducts at N- and O-atoms in DNA bases. These lesions are mainly repaired by direct base repair, base excision repair, and to some extent by nucleotide excision repair (NER). The identified carcinogenicity of O(6)-methylguanine (O(6)-meG) is largely caused by its miscoding properties. Mutations from this lesion are prevented by O(6)-alkylG-DNA alkyltransferase (MGMT or AGT) that repairs the base in one step. However, the genotoxicity and cytotoxicity of O(6)-meG is mainly due to recognition of O(6)-meG/T (or C) mispairs by the mismatch repair system (MMR) and induction of futile repair cycles, eventually resulting in cytotoxic double-strand breaks. Therefore, inactivation of the MMR system in an AGT-defective background causes resistance to the killing effects of O(6)-alkylating agents, but not to the mutagenic effect. Bifunctional alkylating agents, such as chlorambucil or carmustine (BCNU), are commonly used anti-cancer drugs. DNA lesions caused by these agents are complex and require complex repair mechanisms. Thus, primary chloroethyl adducts at O(6)-G are repaired by AGT, while the secondary highly cytotoxic interstrand cross-links (ICLs) require nucleotide excision repair factors (e.g. XPF-ERCC1) for incision and homologous recombination to complete repair. Recently, Escherichia coli protein AlkB and human homologues were shown to be oxidative demethylases that repair cytotoxic 1-methyladenine (1-meA) and 3-methylcytosine (3-meC) residues. Numerous AlkB homologues are found in viruses, bacteria and eukaryotes, including eight human homologues (hABH1-8). These have distinct locations in subcellular compartments and their functions are only starting to become understood. Surprisingly, AlkB and hABH3 also repair RNA. An evaluation of the biological effects of environmental mutagens, as well as understanding the mechanism of action and resistance to alkylating drugs require a detailed understanding of DNA repair processes.
Collapse
Affiliation(s)
- Finn Drabløs
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Katsoulas A, Rachid Z, Brahimi F, McNamee J, Jean-Claude BJ. Engineering 3-alkyltriazenes to block bcr-abl kinase: a novel strategy for the therapy of advanced bcr-abl expressing leukemias. Leuk Res 2005; 29:693-700. [PMID: 15863211 DOI: 10.1016/j.leukres.2004.11.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Accepted: 11/09/2004] [Indexed: 11/30/2022]
Abstract
Recently, within the framework of a new strategy termed "combi-targeting," we designed ZRCM5 to contain a 2-phenylaminopyrimidopyridine moiety targeted to bcr-abl kinase and a triazene tail capable of generating a methyldiazonium species upon hydrolysis. The ability of ZRCM5 to block tyrosine kinase activity was tested in a short 10 min exposure ELISA involving isolated bcr-abl kinase and Western blotting assays. The results showed that: (a) ZRCM5 was hydrolyzed with a half-life of 27 min in cell culture media, (b) it blocked bcr-abl autophosphorylation in promyeloblastic leukemia K562 cells in a dose-dependent manner (IC(50)=14.01 microM) and (c) it induced dose-dependent levels of DNA strand breaks. In contrast, temozolomide (TEM), a clinical DNA damaging triazene capable of generating, like ZRCM5, a methyldiazonium species, could neither block bcr-abl tyrosine kinase activity in isolated enzyme nor in whole cell autophosphorylation assays. In cells expressing varied levels of bcr-abl, ZRCM5 was consistently more potent than TEM. The significant potency of ZRCM5 against the leukemia cells was attributed to its ability to simultaneously to block bcr-abl and related DNA repair activity while inducing significant DNA lesions in bcr-abl expressing leukemia cells. Further studies are ongoing to increase the affinity of ZRCM5 with the purpose of further enhancing its potency in bcr-abl expressing cells.
Collapse
Affiliation(s)
- Athanasia Katsoulas
- Cancer Drug Research Laboratory, Department of Medicine, Division of Medical Oncology, McGill University Health Center/Royal Victoria Hospital, 687 Pine Ave. West, M7.19, Montreal, Que., Canada H3A 1A1
| | | | | | | | | |
Collapse
|
45
|
Shanafelt TD, Lee YK, Bone ND, Strege AK, Narayanan VL, Sausville EA, Geyer SM, Kaufmann SH, Kay NE. Adaphostin-induced apoptosis in CLL B cells is associated with induction of oxidative stress and exhibits synergy with fludarabine. Blood 2004; 105:2099-106. [PMID: 15388586 DOI: 10.1182/blood-2004-06-2205] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (CLL) is characterized by accumulation of clonal lymphocytes resistant to apoptosis. We evaluated the ability of the investigational antileukemic agent adaphostin to induce apoptosis in CLL B cells and synergize with fludarabine in vitro. Analysis by annexin V/propidium iodide (PI) staining revealed that the concentration of adaphostin required to induce 50% cell death (IC50) at 24 hours was 4.2 microM (range, 1.10-11.25 microM; median, 4.25 microM; n=29) for CLL isolates and more than 10 microM for B and T cells from healthy donors. Immunoblots demonstrated adaphostin induced poly(adenosine diphosphate-ribose) polymerase (PARP) cleavage and cleavage of caspase-3 substrates, suggesting that adaphostin induces apoptosis. Adaphostin increased the level of reactive oxygen species (ROS) within CLL B cells, and the antioxidant N-acetylcysteine blocked both adaphostin-induced ROS generation and apoptosis. Adaphostin also caused a decrease in the level of the antiapoptotic protein Bcl-2. When adaphostin was combined with fludarabine (F-ARA-AMP), a synergistic effect on cell death was observed in all 10 CLL samples. These findings not only indicate that adaphostin induces apoptosis selectively in CLL B cells through a mechanism that involves ROS generation but also demonstrate its ability to augment the effects of fludarabine. Further preclinical development of adaphostin as a novel agent for the treatment of CLL appears warranted.
Collapse
Affiliation(s)
- Tait D Shanafelt
- Department of Medicine, Division of Hematology, Mayo Clinic, 200 First St, SW, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|