Clinical, pharmacokinetic (PK), and pharmacodynamic findings from a phase I trial of an Eg5 inhibitor (AZD4877) in patients with refractory acute myeloid leukemia (AML)

2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 3580-3580
Author(s):  
G. Borthakur ◽  
S. Faderl ◽  
F. Ravandi ◽  
S. Padmanabhan ◽  
W. Stock ◽  
...  

3580 Background: AZD4877 is a potent, specific inhibitor of Eg5 (kinesin spindle protein). The only known function of Eg5 is to separate the centromeres during mitosis. Eg5 inhibition is thus specific for dividing cells, resulting in monoastral mitotic spindles (monoasters) and apoptotic cell death. Preclinically, hematologic tumor cell lines were generally more sensitive to AZD4877 than those derived from solid tumors. Methods: AZD4877 was administered IV daily x 3 as induction for up to 2 cycles, followed by consolidation (daily x 2) for up to 4 cycles. Eligibility criteria were standard. Results: Cohorts of 3–6 patients (pts) were treated at doses of 2, 4, 7, 10, 13, 16, and 18 mg/day. To date, 24 evaluable pts have received 33 induction and 3 consolidation cycles of treatment. Monoasters were detected at all dose levels evaluated (2, 7, 10, 13, and 18 mg/day). The T1/2 of AZD4877 ranged from 26 to 42 hr; PK were linear and drug levels non-cumulative. Myelosuppression, the dose limiting toxicity (DLT) in solid tumor studies, was not considered a DLT in this trial. Mucositis was the DLT at 18 mg/day; with 1 pt developing Gr 3 palmar-plantar syndrome at this dose. Bone marrow biopsies were undertaken at screening and as clinically indicated. In pts with evaluable biopsies, marrow blasts decreased by 60–80% in 2 pts, and by 30–50% in 3 pts, with no response in 3 pts. Conclusions: Enrollment is ongoing at 16 mg, the likely Phase 2 dose. Preliminary results suggest possible clinical activity in AML. An expansion phase at the MTD is planned. [Table: see text]

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3838-3838 ◽  
Author(s):  
Jeffrey E Lancet ◽  
Andrew H Wei ◽  
Simon TS Durrant ◽  
Mark S. Hertzberg ◽  
Ronan T. Swords ◽  
...  

Abstract Background EphA3 is involved in cell positioning in fetal development. In the adult it is an oncofetal antigen re-expressed in hematologic malignancies (blood and bone marrow, leukemic stem cells) and solid tumors (tumor stem cells, neovasculature and stroma) and may be prognostic. KB004 is a humaneered® high affinity antibody targeted against EphA3 with 3 putative mechanisms of action: direct induction of apoptosis in tumor cells, activation of ADCC and disruption of tumor vasculature by induction of endothelial cell rounding and subsequent blood vessel infarction. Herein, we describe results of an ongoing, phase 1 dose-escalation study of KB004 in adult patients with advanced hematologic cancers. Objectives 1) primary: to determine safety and MTD for KB004 in patients with hematologic malignancies refractory to or unfit for chemotherapy; 2) secondary: to examine PK profile, immunogenicity, and clinical activity of KB004. Exploratory objectives include evaluation of EphA3 expression on tumor, stromal, and endothelial cells. Methods This is a multicenter phase 1 study. Key eligibility criteria include: 1) relapsed or refractory hematologic malignancy 2) ECOG PS 0-1; 3) adequate end-organ function. Additional eligibility criteria amended later to protect against hemorrhagic events, included platelets ≥ 10,000/uL (untransfused) and normal coagulation times. A standard 3 + 3 escalation study design (amended to allow up to 6 pts. per cohort in the absence of a DLT) was utilized. KB004 was administered as a 1 or 2 hr infusion on days 1, 8, and 15 of each 21-day cycle, at incremental doses of 20, 40, 70, 100, 140, 190, 250 mg and thereafter 33% increments up to a planned maximum of 700 mg. At 70 mg and above infusion reaction (IR) prophylaxis included an H1 blocker, H2 blocker, acetaminophen and IV steroids. Peripheral blood and bone marrow biopsy specimens for PK analysis and EphA3 expression, respectively, were collected during cycle 1. Results To date, 37 patients (AML 32, MDS 2, lymphoma 1, myelofibrosis 2) received KB004; 20 mg: 9 pts, 40 mg: 3pts, 70 mg: 8 pts, 100 mg: 7pts, 140 mg: 5pts, 190 mg 5pts. At 70 mg, two patients had intracranial hemorrhages 5 and 18 days after last KB004 dose in the context of thrombocytopenia and hyperleukocytosis. A causal relationship to KB004 could not be excluded. One occurred in course 1 and was considered a DLT. KB004 blood levels were near the limit of quantitation at 48 and 96 hours. Following the change to entry criteria no further episodes of serious bleeding or other DLTs or significant changes in soluble clotting factors have been observed. Overall KB004 is well tolerated. The most common toxicity was mild to moderate transient IRs in 28 (76%) patients characterized by chills, elevated temperature, fever, rigors, back pain, nausea, vomiting, hypotension, hypertension, transient hypoxia (in 2 cases). Fourteen % of infusions were slowed due to an adverse event, two (1.2%) were prematurely discontinued but no patient discontinued KB004 secondary to an IR. No other significant KB004-related toxicity has been observed. KB004 Cmax at all dose levels was above the predicted effective concentration (1 ug/ml) and was approximately dose proportional. However at dose levels below 190 mg sustained exposure to cover the entire interval between doses was not achieved. One CRp was observed at the 20mg dose level in a 78 yr-old patient with relapsed AML, who remains on study and in sustained remission for over a year. Serial bone marrow biopsies with KB004 treatment show decreased reticulin and collagen fibrosis. A > 50% reduction in marrow blast percentage was seen in 14% of AML patients, and 59% had overall stable disease beyond 1 cycle. Bone marrow biopsies positive for EphA3 expression with a cut-off of 10% of nucleated cells were obtained in 75% of AML patients. EphA3 was expressed in at least 30% of nucleated cells in the baseline sample of the patient with an ongoing CRp. Conclusion KB004 is a novel agent targeted against EphA3 that is well tolerated with evidence of clinical activity. It is anticipated that 190 mg given over 2 hours will provide sufficient plasma exposure to achieve sustained efficacy over the interval between doses. The study is ongoing. Additional PK, PD, and clinical data will be presented. Disclosures: Durrant: KaloBios: Research Funding. Yarranton:KaloBios: Employment, Equity Ownership; Glaxo: Equity Ownership; EnGen: Equity Ownership, Science Advisor, Science Advisor Other; StemLine Therapeutics: Equity Ownership. Walling:KaloBios: Consultancy; Corcept Therapeutics: Consultancy; Prothena: Consultancy; New Gen Therapeutics: Consultancy; Valent Technologies: Consultancy; LBC Pharmaceuticals: Consultancy; Amgen: Equity Ownership; BioMarin: Equity Ownership; Crown BioScience: Membership on an entity’s Board of Directors or advisory committees.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Eun-Ah Park ◽  
Juri Kim ◽  
Mee Young Shin ◽  
Soon-Jung Park

Abstract Background Polo-like kinases (PLKs) are conserved serine/threonine kinases that regulate the cell cycle. To date, the role of Giardia lamblia PLK (GlPLK) in cells has not been studied. Here, we report our investigation on the function of GlPLK to provide insight into the role of this PKL in Giardia cell division, especially during cytokinesis and flagella formation. Methods To assess the function of GIPLK, Giardia trophozoites were treated with the PLK-specific inhibitor GW843286X (GW). Using a putative open reading frame for the PLK identified in the Giardia genomic database, we generated a transgenic Giardia expressing hemagglutinin (HA)-tagged GlPLK and used this transgenic for immunofluorescence assays (IFAs). GlPLK expression was knocked down using an anti-glplk morpholino to observe its effect on the number of nuclei number and length of flagella. Giardia cells ectopically expressing truncated GlPLKs, kinase domain + linker (GlPLK-KDL) or polo-box domains (GlPLK-PBD) were constructed for IFAs. Mutant GlPLKs at Lys51, Thr179 and Thr183 were generated by site-directed mutagenesis and then used for the kinase assay. To elucidate the role of phosphorylated GlPLK, the phosphorylation residues were mutated and expressed in Giardia trophozoites Results After incubating trophozoites with 5 μM GW, the percentage of cells with > 4 nuclei and longer caudal and anterior flagella increased. IFAs indicated that GlPLK was localized to basal bodies and flagella and was present at mitotic spindles in dividing cells. Morpholino-mediated GlPLK knockdown resulted in the same phenotypes as those observed in GW-treated cells. In contrast to Giardia expressing GlPLK-PBD, Giardia expressing GlPLK-KDL was defective in terms of GIPLK localization to mitotic spindles and had altered localization of the basal bodies in dividing cells. Kinase assays using mutant recombinant GlPLKs indicated that mutation at Lys51 or at both Thr179 and Thr183 resulted in loss of kinase activity. Giardia expressing these mutant GlPLKs also demonstrated defects in cell growth, cytokinesis and flagella formation. Conclusions These data indicate that GlPLK plays a role in Giardia cell division, especially during cytokinesis, and that it is also involved in flagella formation.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS9134-TPS9134
Author(s):  
Joel W. Neal ◽  
Palak Kundu ◽  
Tomohiro Tanaka ◽  
Ida Enquist ◽  
Sid Patel ◽  
...  

TPS9134 Background: Patients with mNSCLC who progress on anti–PD-L1/PD-1 therapy administered in combination with or after platinum-based chemotherapy (PBC) are mainly treated with docetaxel or pemetrexed monotherapy. These therapies only have modest clinical activity, leaving a high unmet medical need. Cabozantinib, a tyrosine kinase inhibitor (TKI), promotes an immune-permissive environment and may enhance the efficacy of PD-L1/PD-1 inhibitors, offering a promising second/third-line therapeutic opportunity for patients with mNSCLC. In a Phase Ib multi-cohort study (COSMIC-021; NCT03170960), cabozantinib plus atezolizumab (anti–PD-L1) showed an acceptable safety profile and promising efficacy (ORR: 27%; mDOR: 5.7 mo [range: 2.6-6.9]; disease control rate [CR + PR + SD]: 83%) in 30 patients with mNSCLC who had progressed after prior anti–PD-L1/PD-1 therapy plus chemotherapy (Neal et al. J Clin Oncol 2020). The Phase III CONTACT-01 study will further evaluate the efficacy and safety of atezolizumab plus cabozantinib versus docetaxel monotherapy in patients with mNSCLC who have progressed during or after prior treatment with anti–PD-L1/PD-1 therapy and PBC. Methods: CONTACT-01 (NCT04471428) is a Phase III, multi-center, randomized, open-label study that will enroll ≈350 patients from 150 to 200 sites internationally. Key eligibility criteria include histologically or cytologically confirmed mNSCLC, disease progression with concurrent or sequential anti–PD-L1/PD-1 treatment and PBC, measurable disease (RECIST 1.1), ECOG PS of 0-1 and the availability of tissue specimens for centralized PD-L1 testing or known PD-L1 status using a health authority–approved PD-L1 assay. Patients with NSCLC previously treated with cabozantinib, docetaxel or anti–PD-L1/PD-1 + VEGFR TKIs are excluded. Patients with known sensitizing EGFR/ALK mutations and active or untreated CNS metastases are also excluded. Patients will be randomized 1:1 to receive either atezolizumab (1200 mg IV every 3 weeks) + cabozantinib (40 mg orally once daily) or docetaxel (75 mg/m2 IV every 3 weeks). The primary endpoint is OS. Secondary endpoints include investigator-assessed PFS, ORR and DOR per RECIST 1.1; TTD in patient-reported physical function and global health status (EORTC QLQ-C30); investigator-assessed PFS rates at 6 months and 1 year; OS rates at 1 and 2 years; safety and PK. Clinical trial information: NCT04471428.


2004 ◽  
Vol 279 (28) ◽  
pp. 29728-29739 ◽  
Author(s):  
Dana R. Rundle ◽  
Gary Gorbsky ◽  
Leonidas Tsiokas

1998 ◽  
Vol 4 (S2) ◽  
pp. 1036-1037
Author(s):  
M. C. Willingham

Several clinically important anti-cancer agents exert their effects on tumor cells through interference with the function of microtubules. In addition to the Vinca alkaloids, such as vinblastine and vincristine, the taxanes, such as paclitaxel (Trade Name: Taxol), kill tumor cells through a microtubular target. Treatment with taxol leads to the inability of microtubules to depolymerize, leading to the formation of large intracellular microtubular bundles. In tumor cells that progress through the cell cycle, this leads to the inability of these cells to disassembly interphase microtubule networks and a failure to form functional mitotic spindles. These cells arrest in M phase, from which they eventually progress, either by the induction of apoptotic cell death, or by micronucleation and the formation of tetraploid cells. There is also the possibility that taxol has other effects on the regulation of genes or other systems to enhance cell killing, perhaps through lowering the threshold of cells to the induction of apoptotic cell death.


2005 ◽  
Vol 280 (43) ◽  
pp. 36502-36509 ◽  
Author(s):  
Fumihiko Niiya ◽  
Xiaozhen Xie ◽  
Kyung S. Lee ◽  
Hiroki Inoue ◽  
Toru Miki

Cleavage furrow formation marks the onset of cell division during early anaphase. The small GTPase RhoA and its regulators ECT2 and MgcRacGAP have been implicated in furrow ingression in mammalian cells, but the signaling upstream of these molecules remains unclear. We now show that the inhibition of cyclin-dependent kinase (Cdk)1 is sufficient to initiate cytokinesis. When mitotically synchronized cells were treated with the Cdk-specific inhibitor BMI-1026, the initiation of cytokinesis was induced precociously before chromosomal separation. Cytokinesis was also induced by the Cdk1-specific inhibitor purvalanol A but not by Cdk2/Cdk5- or Cdk4-specific inhibitors. Consistent with initiation of precocious cytokinesis by Cdk1 inhibition, introduction of anti-Cdk1 monoclonal antibody resulted in cells with aberrant nuclei. Depolymerization of mitotic spindles by nocodazole inhibited BMI-1026-induced precocious cytokinesis. However, in the presence of a low concentration of nocodazole, BMI-1026 induced excessive membrane blebbing, which appeared to be caused by formation of ectopic cleavage furrows. Depletion of ECT2 or MgcRacGAP by RNA interference abolished both of the phenotypes (precocious furrowing after nocodazole release and excessive blebbing in the presence of nocodazole). RNA interference of RhoA or expression of dominant-negative RhoA efficiently reduced both phenotypes. RhoA was localized at the cleavage furrow or at the necks of blebs. We propose that Cdk1 inactivation is sufficient to activate a signaling pathway leading to cytokinesis, which emanates from mitotic spindles and is regulated by ECT2, MgcRacGAP, and RhoA. Chemical induction of cytokinesis will be a valuable tool to study the initiation mechanism of cytokinesis.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1509-1509 ◽  
Author(s):  
Ruben A. Mesa ◽  
John K. Camoriano ◽  
Susan M. Geyer ◽  
Scott H. Kaufmann ◽  
Candido E. Rivera ◽  
...  

Abstract Background: Myelofibrosis with myeloid metaplasia (MMM) is a progressive and fatal myeloproliferative disorder with limited therapeutic options. R115777 is a non-peptidomimetic enzyme-specific inhibitor of farnesyl protein transferase (FT) which we recently reported has in vitro activity against aberrant myeloid progenitors from MMM patients (Leukemia2003;17:849). This observation, coupled with the clinical activity demonstrated in high risk acute myeloid leukemia (Blood2001;97:3361) and chronic myeloid disorders (Blood2003;101:1692) led us to perform a Phase II trial in patients (pts) with MMM. Methods: Eligible pts had histologically confirmed MMM and were symptomatic, defined by anemia (hemoglobin<10g/dL or transfusion dependent) or palpable hepatosplenomegaly. Pts received 300 mg of R115777 orally twice daily for the first 21 days of a 28 day cycle. Dose escalations (up to 1200mg/day) and reductions (down to 100mg/day) were allowed based on activity and toxicity. The primary endpoint was response as defined by improvement in either anemia or organomegaly. Results: 34 pts (median age 65 yrs, range 55–80; 71% male) were enrolled in the trial. All pts displayed symptomatic disease at enrollment, with 10 who were erythrocyte transfusion dependent; and 19 (56%) had a Lille MMM prognostic score ≥1; no pts were significantly thrombocytopenic at entry by enrollment criteria. Median pre-study spleen and liver sizes were 12 cm (range: 0–35) and 2 cm (range: 0–15) below the left and right costal margin, respectively. Median time to discontinuation of therapy was 5.5 months (95% CI:2.9–6.4). Reasons for early termination were progression (7), adverse reactions (5), co-morbidities (2), refusal of further treatment (2), and death on study (1). One pt died of grade 5 pneumonitis two weeks after study enrollment, which was felt to be unrelated to study treatment. Significant toxicities (≥ grade 3) were seen for myelosuppression (n=13), neuropathy (n=2), fatigue (n=1), rash (n=1) and hyponatremia (n=1). Overall, R115777 resulted in little improvement in anemia, with the exception of one anemia-based partial response. However, the myelosuppressive aspects of the agent may have masked improvements in cytopenias. In contrast, R115777 did result in clinically relevant decreases in organomegaly in 11 pts (33%) (splenomegaly in 3 pts, hepatomegaly in 5 pts, and both in an additional 3 pts.) Across all pts who had some reduction in splenomegaly, the median maximal reduction was 42.5% (of palpable component; range: 8–100%). Likewise, all patients with baseline hepatomegaly had resolution of this problem at some point during therapy. Four pts remain on the agent (from 14 to 22 months) for continued and sustained improvement in organomegaly. Responses observed did not significantly correlate with reductions in bone marrow fibrosis, osteosclerosis, neoangiogenesis, or resolution of baseline karyotypic abnormalities. Conclusions: R115777 has demonstrated preliminary activity against MMM associated organomegaly. Whether this latter benefit is independent of R115777 induced myelosuppression is unclear. The myelosuppression observed was generally mild and without clinical consequence, although it was difficult to differentiate between myelosuppression versus disease progression. Pts continue to be followed to evaluate durability of observed responses, long term effects on anemia and intramedullary manifestations of the disease.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5258-5258
Author(s):  
Annie Levesque ◽  
Ann-Louise Savard ◽  
Denis-Claude Roy ◽  
Francine Foss ◽  
Christian Scotto

Abstract Recently, extracorporeal photopheresis (ECP) has shown interesting clinical activity for the treatment of drug-refractory chronic graft-vs-host disease (cGvHD), inducing Th1/Th2 immunomodulation to restore immune tolerance. Several studies indicate that target cell apoptosis may have a role in the control of cGVHD, and increasing apoptotic levels may favor immune modulation. We have developed an approach to eliminate immunoreactive cells using the Theralux™ photodynamic cell therapy (PDT) system based on the use of the rhodamine-derivative TH9402 illuminated ex vivo with a visible light source (l=514nm). The capacity of TH9402 PDT to induce increasing levels of T cell apoptosis has not been investigated. The induction of apoptotic cell death was studied using peripheral blood mononuclear cells (PBMCs) isolated from healthy volunteers (HV) and cGvHD patients. We found a good correlation between PDT conditions and levels of cell death induced. In comparison to controls showing 8±4 percent (%) of apoptosis, as measured by TUNEL assay on cells harvested 3 days post-treatment, PBMC from HV subjected to PDT using 0.33, 0.66, or 1.32 micromolar of TH9402 showed 28±16%, 49±17%, and 78±11% of apoptosis, respectively. These studies have also shown that the intra- and inter-donor variability in TH9402 incorporation are very low (~5% and 10%, respectively). To ensure that these findings could also be applied to a clinical setting, PBMC from cGvHD patients were treated with 0.33 and 1.32 micromolar of TH9402 to trigger either low or high levels of apoptosis. PBMC from cGvHD patients showed a sensitivity similar to that of PBMC from HV, with 38±9% and 73±13% of apoptosis when treated with 0.33 and 1.32 micromolar TH9402, respectively. Data obtained from the analysis of various cell death parameters such as the loss of DΨm, AnV/PI staining, TUNEL assay and agarose DNA ladder on PBMC and human Jurkat T cells treated with escalating doses of TH9402 suggested strongly the higher sensitivity of proliferating cells to the treatment. Moreover, together with the use of the caspase-inhibitor ZVAD-fmk, these data suggested that upon irradiation, the photoactivation of TH9402 will trigger the formation of reactive oxygen species (ROS) and the release of proapoptotic factors from mitochondria triggering various cell death mechanisms, such as caspase-dependent apoptosis, caspase-independent apoptosis, or a mixture of apoptosis and necrosis. Finally, preliminary data showed that PDT-treated cells were able to induce in vitro the maturation of monocyte-derived dendritic cells. Based on these data, we are beginning a pilot clinical study evaluating two controlled PDT conditions inducing low and high levels of apoptosis in order to assess the efficacy and biological effect of TH9402-based ECP to treat cGVHD in humans.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3448-3448
Author(s):  
Amit Verma ◽  
Tony A. Navas ◽  
Jing Ying ◽  
Aaron N. Nguyen ◽  
Perry Pahanish ◽  
...  

Abstract Transforming Growth Factor β (TGF-β) is a myelosuppressive cytokine that has been implicated in the ineffective hematopoiesis seen in myelodysplastic syndromes (MDS). Overactivation of TGF-β signaling in this disease was demonstrated immunohistochemically by significantly higher nuclear SMAD2 phosphorylation observed in 20 MDS bone marrows when compared with 7 non MDS anemic controls (P &lt; 0.0001, 2 Tailed T Test, Image Pro Plus software). This data along with high levels of membrane-bound and plasma TGF-β observed in MDS patients in previous studies support the development of therapeutics targeting the TGF-β signaling pathways in this disease. SD-208 is a novel, potent and specific inhibitor of TGF-β Receptor I (TGFβ-RI) kinase. We demonstrate that SD-208 blocks the phosphorylation of SMAD2 in hematopoietic progenitors which are at the colony forming unit-erythroid (CFU-E) stage of differentiation. SD-208 also abrogates the G0/G1 cell cycle arrest induced by TGF-β in bone marrow progenitors. SD-208 treatment leads to reversal of the myelosuppressive effects of TGF-β on erythroid and myeloid colony formation from primary human CD34+ cells. Selectivity of SD-208 in inhibiting TGF-β-mediated effects on hematopoiesis was supported by similar results observed with siRNAs targeting SMAD2, a major component of the TGF-b signaling pathway. Finally, the efficacy of SD-208 in MDS was evaluated by treating bone marrow mononuclear cells from 15 patients with early low grade MDS. SD-208 treatment led to dose-dependent increases in erythroid and myeloid colonies after 14 days of in vitro culture. The effect was most notable in patients with high levels of activated SMAD-2, as assessed by immunohistochemical staining of bone marrow biopsies. Stimulation of hematopoiesis in MDS-derived marrow culture by SD-208 demonstrates a novel concept and potential therapeutic role for TGFβ-RI inhibition in this disease. Supported by VISN-17 grant, Harris Methodist Foundation Grant and ASCO YIA to AV


Sign in / Sign up

Export Citation Format

Share Document