scholarly journals Differential expression of aurora-A kinase in T-cell lymphomas

2013 ◽  
Vol 26 (5) ◽  
pp. 640-647 ◽  
Author(s):  
Rashmi Kanagal-Shamanna ◽  
Norman L Lehman ◽  
James P O'Donnell ◽  
Megan S Lim ◽  
Daniel S Schultz ◽  
...  
Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1744-1744 ◽  
Author(s):  
Michelle A. Fanale ◽  
Fredrick B. Hagemeister ◽  
Luis Fayad ◽  
Yasuhiro Oki ◽  
Nathan Fowler ◽  
...  

Abstract Background: The histone deacetylase inhibitor (HDACi) romidepsin, while a clear advance for relapsed peripheral and cutaneous T cell lymphomas (CTCL and PTCL), induces short durations of remission at 9 to 10 months (Piekarz et al., 2011 and Coiffier et al., 2011). Other HDACi have been evaluated in other lymphoma types including Hodgkin lymphoma (HL) with panobinostat having a 27% ORR in patients with post-ASCT relapsed disease (Younes et al., 2012). The aurora A kinase inhibitor alisertib has shown promising results to date including in a phase II sponsored trial (Friedberg et al., 2011) in which the ORR was 32% with responses of 100% in Burkitt lymphoma (BL), 20% in diffuse large B cell lymphoma (DLBCL), and 57% in PTCL. Recent data from a SWOG further showed an ORR of 20% in all TCL and 50% in PTCL (Barr et al., 2014), and a registration trial is ongoing in relapsed PTCL. Preclinical data supports the combination of an aurora A kinase inhibitor plus a HDACi. The pan-aurora kinsase inhibitor MK-0457 in combination with the HDACi vorinostat enhanced lymphoma cell death through repression of C-Myc and C-Myc responsive micro RNAs (Kretzner et al., 2008). Also alisertib plus romidepsin exhibit highly synergistic effects in lymphoma cell lines (O’Connor, 2012). Thus, this collective data supports the rationale for the evaluation of the combination of romidepsin plus alisertib in patients with multiple lymphoma subtypes. Methods: Eligible histologies included Hodgkin lymphoma (HL), Burkitt lymphoma (BL), double-hit lymphoma (DHL), other c-Myc positive B-cell lymphomas, diffuse large-B cell lymphoma (DLBCL), mantle cell lymphoma (MCL), or peripheral T-cell lymphoma (PTCL). Patients were treated with alisertib orally on days 1 to 7 and romidepsin IV on days 1 and 8. There are 5 planned escalation dose levels with respective dosing of alisertib plus romidespin of 20 mg BID and 8 mg/m2, 20 mg BID and 10 mg/m2, 40 mg BID and 10 mg/m2, 40 mg BID and 12 mg/m2, and 40 mg BID and 14 mg/m2. Next cycle is given if ANC ≥ 1000 and platelets ≥ 50,000 and maximum cycles is 8. Restaging is done after every 2 cycles with revised response criteria (Cheson et al., 2007). DLT is defined as: 1) grade 4 neutropenia or thrombocytopenia ≥ 14 days and/or 2) grade 3 or 4 non-hematological toxicity attributed to study drugs that could not be controlled by supportive care. Patients with an ANC < 1000 received growth factor support. A lymph node core biopsy is conducted at baseline and at the end of 1 cycle of therapy, and whole peripheral blood is also collected. Evaluation of intensity of immnohistochemistry (IHC) expression of aurora A kinase will be performed and will be correlated with response, 2. Gene expression profiling (GEP) will be performed and assessments of markers of apoptosis and mitotic catastrophe, 3. GEP of whole peripheral blood will be performed to assess changes beyond those limited to within the tumor that can contribute towards response to therapy. Results: 9 patients were enrolled and 8 are evaluable for response. The median age was 60 years and histologies were 3 PTCL, 3 DHL defined by FISH, 1 DLBCL with c-Myc translocation by FISH, 1 high-grade (HG) DLBCL, and 1 transformed DLBCL. Median number of prior therapies was 4 (2 to 7) and no patients underwent prior transplant given refractory disease. 3 patients have been enrolled to each of the dose levels 1, 2, and 3. Median number of cycles is 1.5 (1 to 8) with median time for retreatment of 28.5 days (22 to 40). Grade 3/4 toxicities were neutropenia, thrombocytopenia, and anemia in respectively 45%, 45%, and 20% of the cycles. Responses to date are CR (PTCL, dose level 1), SD (PTCL, dose level 3), PD (3 DHL, 1 HG DLBCL, 1 DLBCL with c-Myc, 1 PTCL). 4 of the patients with PD have died from continued refractory disease and 1 has been transitioned to hospice. The CR patient received 7 prior lines of treatment and remains in remission at 5 months in follow-up and declined transplant. The SD patient is now 1 month out from a matched unrelated donor transplant. Conclusions: Enrollment continues. Based on preclinical data, clinical data for both agents, and responses thus far we plan to consider a dose expansion PTCL patient cohort at the MTD. Reversible cytopenias are the main toxicity to date. We anticipate the correlative studies will allow us to further define the patients with the higher likelihood of having disease response to this targeted therapeutic combination. Disclosures Fanale: Seattle Genetics: Consultancy, Honoraria, Research Funding; Millennium/Takeda: Honoraria, Research Funding; Celgene: Research Funding; Novartis: Research Funding; Spectrum: Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Research Funding; MedImmune: Research Funding; Roche: Research Funding; Amgen : DMC, DMC Other. Off Label Use: Will discuss off label use of alisertib plus romidepsin in a phase I trial.. Fowler:Gilead Sciences: Research Funding.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 95-95 ◽  
Author(s):  
Jonathan Friedberg ◽  
Daruka Mahadevan ◽  
JungAh Jung ◽  
Daniel O. Persky ◽  
Izidore S Lossos ◽  
...  

Abstract Abstract 95 Background: Aurora kinases are a family of oncogenic serine-threonine kinases that regulate multiple phases of the mitotic signaling cascade. Inhibition of aurora A kinase (AAK) leads to mitotic errors, followed by aneuploidy, apoptosis, and senescence. Investigational drug alisertib is an ATP-competitive, orally available inhibitor of AAK, that has been evaluated for safety and efficacy in Phase I hematological malignancies. We conducted the first phase II multicenter trial of alisertib in adult patients with aggressive B- and T- cell NHL. Methods: Eligible patients had normal organ function, ANC ≥1250/mm3, platelets ≥ 75,000/mm3 and no prior allogeneic transplant. Patients were treated with alisertib at a dose of 50mg twice daily for 7 days on 21 day cycles until either documented progression or unacceptable treatment-related toxicity. Using fluorescent in situ hybridization (FISH), gene amplification was assessed in archived tumors by a dual assay measuring AAK copy number on chromosome 20q13 as well as the ratio to a control probe located on chromosome 20q11. Immunohistochemistry (IHC) was performed on archived paraffin embedded diagnostic tissue using a dual assay measuring the protein levels of total AAK and that of phospho histone H3. PK sampling was performed with inclusion of steady-state trough plasma PK samples on the morning of Cycle 1 Day 8. Results: 48 pts were enrolled, including 41 response-evaluable. Histologies included DLBCL (n=21, 44%), mantle cell (MCL; n=13, 27%), peripheral T- cell (n=8, 17%), transformed follicular (n=5, 10%) and Burkitt (n=1, 2%). Median age was 68 y (range 32–85). Pts received median 3 prior regimens (range 1–11); 11 pts received prior ASCT. Most common Grade 3/4 adverse events were neutropenia (63%), thrombocytopenia (31%), stomatitis (15%), febrile neutropenia (13%) and fatigue (6%). Four deaths on study were attributed to progressive NHL (2), treatment-related sepsis (1), and unknown cause (1). 11 pts discontinued and 24 reduced dose due to adverse events (AEs). Evaluation of steady state trough concentration of alisertib (N=25) revealed that PK variability was consistent with that observed in other trials with alisertib, and there was a trend toward higher levels in patients who required AE-related dose reductions compared to those who did not (geometric mean 2375 nM [n=10, CV: 54%] vs. 1504 nM [n=15, CV: 35%]). FISH analysis for AAK gene amplification did not reveal differences between histologies (N=31). When total AAK protein was evaluated by immunohistochemistry (N=32), marked variability in both proportion of expression as well as intensity was observed both between and within histologies; there was no correlation of AAK protein expression and clinical response. The overall response rate (ORR) was 32% (95% CI 0.181–0.481); response by histology: DLBCL 20%; MCL 23% and T- cell NHL 57%. Conclusion: Current data suggests that alisertib is generally well-tolerated, with responses observed in heavily pretreated patients with aggressive NHL, including patients after ASCT. Emerging data supports single agent activity in several histologies, with proportionally more responses observed in relapsed/refractory T-cell NHL. Some patients have now been treated for up to two years with this agent, and the generally manageable toxicity profile suggests an opportunity to combine this drug with other agents. IHC of aggressive lymphoma histologies suggests heterogeneity in AAK protein expression and intensity. The geometric mean of alisertib steady-state trough concentration was 1.8 μM (CV= 47%, N = 25), which was above the 1 μM steady-state plasma concentrations associated with saturating levels of pharmacodynamics and antitumor activity in preclinical xenograft models. The observed trend for association between trough concentration of alisertib and AEs supports the dose modification scheme implemented in this trial. Based upon these results, planned future trials include a single-agent study in T-cell NHL, and a combination study exploring alisertib with rituximab and vincristine in aggressive B-cell NHL. Disclosures: Friedberg: Genentech: Consultancy; astellas:; Lilly:; Abbott/Trubion:; Seattle Genetics: Honoraria; Cephalon: Consultancy. Off Label Use: novel agents for relapsed DLBCL. Jung:Millennium: Employment. Danaee:Millennium Pharmaceuticals Inc.: Employment. Zhou:millennium: Employment. Leonard:millennium: Employment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4290-4290
Author(s):  
Kozo Nagai ◽  
Hiroshi Fujiwara ◽  
Toshiki Ochi ◽  
Jun An ◽  
Toshiaki Shirakata ◽  
...  

Abstract Abstract 4290 Background & Purpose: Recently we have identified a novel HLA-A*0201-restricted antigenic 9mer epitope (aa207-215: YLILEYAPL) derived from Aurora-A kinase (Aurora-A) which is capable of generating anti-leukemia cytotoxic T-lymphocytes (CTL).(Blood, 2009) To improve the feasibility of Aurora-A targeting cellular immunotherapy against leukemia, we have established a Aurora A207-215-specific CTL clone, and have obtained the full-length T-cell receptor (TCR) α/β genes for TCR gene transfer. In this study, using human leukemia cell lines and patients' leukemia cells, we examined the anti-leukemia reactivity of engineered T-cells with Aurora-A-specific TCR gene transfer. Methods: Full-length of an HLA-A*0201-restricted and Aurora-A207-215-specific TCR α/β genes (Vα3/J20/Cα, Vβ10.3(12)/J1.1/Cβ1, respectively) were cloned into bicistronic GaLV-pseudotyped retroviral vector. Using Retronectin (Takara Bio. Japan)-coated plates, Aurora-A-specific TCR α/β genes were inserted into lymphocytes. Whether this vector was capable of generating a functional Aurora-A207-215-specific TCR heterodimer was examined using Jurkat/MA cells (kindly gifted from Prof. Erik Hooijberg, Netherlands). The epitope-specific and leukemia specific cytotoxicity and IFN-γ production of gene-modified normal CD8+ and CD4+ T-cells were examined by 51Cr-releasing assay and ELISA. HLA-A*0201-restriction of engineered T-cell responses was examined by inhibition assay with antibodies, and HLA-A*0201 transduced human leukemia cell line: MEG01 which abundantly expresses Aurora-A. In vivo anti-leukemia effect of gene-modified CD8+T-cells was examined using NOD/SCID/γcnull (NSG) mice. Eventually, the on-target adverse effect of these Aurora-A-specific TCR-gene transferred CD8+T-cells against autologous hematopoietic progenitor cells was examined using cord blood CD34+ cells. Results: The Aurora-A specific-TCR expressing retroviral vector was capable of generating a functional TCR in Jurkat/MA cells which could produce luciferase in response to Aurora-A peptide on C1R-A2 cells in a dose dependent manner. Aurora-A-specific TCR-transduced CD8+ T-cells produced IFN-γ and exerted cytotoxicity against Aurora-A peptide-loaded C1R-A2 cells in an HLA-A*0201 restricted fashion. These engineered CD8+ T-cells also killed HLA-A*0201+ leukemia cell line and patient leukemia cells, but not HLA-A*0201+ normal PBMC and normal mitotic PHA-stimulated lymphoblasts. The anti-leukemia effect of These engineered CD8+ T cells was significantly abrogated by the anti-HLA-class I monoclonal antibody (MoAb), but not by anti-HLA-DR-MoAb. These engineered CD8+ T-cells killed HLA-A*0201-transduced MEG01 cells which were abundantly expressing Aurora-A, but not parent HLA-A*0201-negative MEG01 cells. Aurora-A-specific TCR gene transduced CD4+ T-cells produced IFN-γ in response to the epitope recognition, which was also in an HLA-A*0201-restricted fashion. Furthermore, Aurora-A-specific TCR-transduced CD8+ T cells did not damage the viability of autologous cord blood CD34+ cells in vitro. Finally, These engineered CD8+ T-cells successfully inhibited the engraftment and growth of inoculated leukemia cell line cells in the NOD/SCIDγ/cnull mice. Background: In this study, Aurora-A kinase-specific TCR gene transferred T-cells successfully recognized the target epitope and exerted the target-specific cytotoxicity. Additionally these engineered CD8+ T-cells exerted anti-leukemia effect both in vitro and in vivo. While those these transfectants did not damage autologous hematopoietic progenitor cells in vitro. Collectively, the novel anti-leukemia adoptive therapy using Aurora-A-specific TCR-gene transferred T-cells appears promising, and further investigations are warranted for the clinical application. Disclosures: No relevant conflicts of interest to declare.


2003 ◽  
Vol 120 (6) ◽  
pp. 866-873 ◽  
Author(s):  
David M. Dorfman ◽  
Peter van den Elzen ◽  
Andrew P. Weng ◽  
Aliakbar Shahsafaei ◽  
Laurie H. Glimcher

2018 ◽  
Vol 89 (1) ◽  
pp. 88-91 ◽  
Author(s):  
George Jour ◽  
Phyu P. Aung ◽  
E. Dean Merrill ◽  
Jonathan L. Curry ◽  
Michael T. Tetzlaff ◽  
...  

2019 ◽  
Vol 3 (11) ◽  
pp. 1778-1787 ◽  
Author(s):  
Carlos Murga-Zamalloa ◽  
Kedar V. Inamdar ◽  
Ryan A. Wilcox

Abstract High-risk lymphomas (HRLs) are associated with dismal outcomes and remain a therapeutic challenge. Recurrent genetic and molecular alterations, including c-myc expression and aurora A kinase (AAK) and polo-like kinase-1 (PLK1) activation, promote cell proliferation and contribute to the highly aggressive natural history associated with these lymphoproliferative disorders. In addition to its canonical targets regulating mitosis, the AAK/PLK1 axis directly regulates noncanonical targets, including c-myc. Recent studies demonstrate that HRLs, including T-cell lymphomas and many highly aggressive B-cell lymphomas, are dependent upon the AAK/PLK1 axis. Therefore, the AAK/PLK1 axis has emerged as an attractive therapeutic target in these lymphomas. In addition to reviewing these recent findings, we summarize the rationale for targeting AAK/PLK1 in high-risk and c-myc–driven lymphoproliferative disorders.


2007 ◽  
Vol 21 (1) ◽  
pp. 59-64 ◽  
Author(s):  
L.A. NOORDUYN ◽  
R.C. BELJAARDS ◽  
S.T. PALS ◽  
P. VAN HEERDE ◽  
T. RADASZKIEWICZ ◽  
...  

Leukemia ◽  
2005 ◽  
Vol 19 (12) ◽  
pp. 2254-2263 ◽  
Author(s):  
B Martínez-Delgado ◽  
◽  
M Cuadros ◽  
E Honrado ◽  
A Ruiz de la Parte ◽  
...  

2015 ◽  
Vol 21 (18) ◽  
pp. 4097-4109 ◽  
Author(s):  
Kelly M. Zullo ◽  
Yige Guo ◽  
Laurence Cooke ◽  
Xavier Jirau-Serrano ◽  
Michael Mangone ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document