A Phase I, Multi-Center, Dose Escalation Study of Atiprimod in Patients with Refractory or Relapsed Multiple Myeloma (MM).

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 111-111 ◽  
Author(s):  
Michael Wang ◽  
Moshe Talpaz ◽  
Sundar Jagannath ◽  
Asher Alban Chanan-Khan ◽  
Raymond Alexanian ◽  
...  

Abstract Atiprimod (N-N-diethl-8, 8-dipropyl-2-azaspiro [4,5] decane-2-propanamine) is an orally bioavailable cationic amphilic compound that inhibited STAT 3 activation in MM cells. It effectively blocked the signaling pathway of interleukin-6, resulting in activation of caspase 3 and apoptosis (Amit-Vazina et al, Br J Cancer, 2005). Atiprimod has also induced cytotoxicity in dexamethasone, doxorubicin, and melphalan resistant MM cell lines (Hamasaki et al, Blood, 2005). Based on these encouraging in vitro data, we initiated a multi-center, phase I trial of atiprimod for patients (pts) with refractory or relapsed MM who had 2 prior lines of therapy and serum creatinine less than 2 mg/dl. Primary objectives were to evaluate the safety of atiprimod in MM pts and to identify the maximum tolerated dose (MTD). Each cycle of treatment consisted of 14 consecutive days of oral atiprimod followed by 14 consecutive days without treatment. A standard phase I dose escalation was used to determine MTD with atiprimod dose levels at 30 mg, 60 mg, 90 mg, 120 mg, and 180 mg. To date, 14 pts from 4 centers have been enrolled with evaluable data in 12 patients. Median age was 60 (range 44–64); median prior lines of therapy were 4 (range 3–7); median duration from initial treatment to registration to this trial was 36 months (range 19–76). Cohorts of 3 patients have been treated at 30, 60, 90,120 mg/day and 2 patients have been enrolled at the 180 mg/day level; no cohorts have been expanded because of dose-limiting toxicity. Median number of cycles received by MM pts was 2 (range 1–5). Common Grade 1 toxicity events included diarrhea, liver enzyme elevation and dyspepsia. There were two Grade 2 toxicity events with 1 neutropenia at the 90 mg/day level and 1 diarrhea at the 120 mg/day level. One pt had Grade 3 transaminase elevation (peak AST 402, ALT 469 units/L, bilirubin 0.5 mg/dl) during the second cycle that resolved on its own during the 14 day period off treatment. Two patients with rapidly rising serum M proteins prior to enrollment had a transient but clear reduction of their M proteins (30% and 80%) after the first 14 days of atiprimod. Two pts at higher dose levels noted subjective improvement in their bone pain. Atiprimod was generally well tolerated in this heavily treated group of MM pts. The MTD has not been reached. Although there has been no response to date, clinical activity is not expected until higher dose levels are evaluated (240 mg/day, 300 mg/day, and 360 mg/day). After the MTD has been established, the study of atiprimod combinations should be considered based on the in vitro assessment of synergy with other active agents.

2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 6055-6055 ◽  
Author(s):  
P. H. Morrow ◽  
B. S. Glisson ◽  
L. E. Ginsberg ◽  
S. M. Lippman ◽  
M. S. Kies ◽  
...  

6055 Background: Despite recent advances in therapy, patients (pts) with recurrent or metastatic HNSCC continue to demonstrate a poor median survival. In these pts, early trials with pemetrexed, a novel antimetabolite that acts upon several enzymes involved in pyrimidine and purine synthesis, have demonstrated promising efficacy and tolerability. Prior studies found that the administration of oral dexamethasone with pemetrexed reduced the incidence of skin rash. Later, vitamin supplementation (B12 and folic acid), given in addition to the dexamethasone, further diminished side effects. However, no trial has yet evaluated the appropriate steroid dose and its relation to the dosing of pemetrexed, in the setting of vitamin supplementation. We conducted a phase I trial to determine the maximum tolerated dose, toxicity, and preliminary efficacy of pemetrexed when given with different schedules of, or in the absence of, dexamethasone in pts with advanced HNSCC who had been treated with at least one or more chemotherapy regimens. Methods: Eligible pts had metastatic or recurrent HNSCC, prior treatment with one or more chemotherapy regimens, ECOG PS =2, and life expectancy >3 months. A conventional algorithm-based dose escalation design was applied, with three predefined dose levels (DL) of pemetrexed (500 mg/m2, 600 mg/m2, and 700 mg/m2) within each schedule of dexamethasone (none, 20 mg IV on day 1, and 4 mg orally bid for 3 days). Results: A total of 23 pts have been enrolled; 18 pts were evaluable. Median age was 57 years (range 47–82). Median ECOG PS was 1 (range 0–2), and 75% of pts were male. Number of prior chemotherapy regimens were as follows: 1 (40%), 2 (35%), 3 (15%), and 4 (10%). Preliminary data demonstrated only 2 treatment-related adverse events that were grade 3 or greater: anemia (DL1) and pneumonia (DL 1). In all, 13 pts have received pemetrexed with less than standard recommended dexamethasone dosing (none or IV), including 7 pts who received no dexamethasone. Of the 18 evaluable pts, 1 pt had a partial response and 2 pts had stable disease. Conclusions: This represents the first study that demonstrates that steroids may not be required as premedication with pemetrexed. Due to the limited toxicity observed, trial enrollment continues with dose escalation. [Table: see text]


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 6577-6577 ◽  
Author(s):  
Gail J. Roboz ◽  
Hanna Jean Khoury ◽  
Jamile M. Shammo ◽  
Mary Syto ◽  
Francis Burrows ◽  
...  

6577 Background: TG02 is a novel multikinase inhibitor with a unique spectrum of activity, targeting both the cell cycle regulatory cyclin-dependent kinases (CDKs) 1 and 2 and the transcriptional regulators CDKs 7 and 9. TG02 also inhibits the emerging oncogenic MAPK ERK5 and the DNA damage response mediator CDK5. TG02 kills primary blasts from a variety of hematologic cancers and is curative in the MV4-11 model of FLT3-mutant AML. Methods: This is a first-in-man,single arm, open label, phase I dose escalation trial. The primary endpoints are dose-limiting toxicity (DLT), maximally tolerated dose (MTD ) and recommended phase 2 dose (RP2D). Patients (pts) ≥ 18 years with advanced hematological malignancies or newly diagnosed AML pts ≥ 65 years unfit for intensive therapy were enrolled onto daily (A) and intermittent (B, 5 days on 2 days off X 2 weeks) schedules. Pts had acceptable organ function and ECOG PS 1-2. Definition of DLT was G3-4 AST or ALT ≥7 days, G4 AST or ALT, G4 hyperbilirubinemia, any other NCI CTC G3-4 events not due to underlying disease. Dose levels on arm A were 10mg to 70mg and 15mg-150 mg on arm B. Results: Forty-five pts have received at least one dose of study drug. Median age was 66 years (range, 37-87) and 80% were ECOG 0-1. Disease types enrolled included: AML (80%), high-risk MDS (22%), and CML-BC (3%). The median number of previous regimens was 3 (range, 1-12). The MTD on arm A was defined at 50 mg daily based on 2 DLTs at the 70mg dose level (G4 hyperbilirubinemia, G4 fatigue). Enrollment to arm B has competed dose levels 15 (N=3), 30 (N=3), 50 (N=3), 70 (N=3), 100mg (N=3), and enrollment at 150mg is ongoing without DLT to date. Common drug related adverse events were nausea (42%), vomiting (23%), fatigue (18%), decreased appetite (15%), constipation and diarrhea (13% each). Preliminary PK demonstrated dose proportional increases in exposure and a T1/2 , supporting once daily dosing. Conclusions: The MTD for TG02 has been determined for the daily schedule at 50mg. Enrollment continues on the intermittent schedule. Schedules of every other day and week on/week off dosing will also be evaluated.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e16517-e16517 ◽  
Author(s):  
Christos Kyriakopoulos ◽  
Channing Judith Paller ◽  
Ajit Verma ◽  
Karim Kader ◽  
Jeff Kittrelle ◽  
...  

e16517 Background: The combination of PCUR-101 (a synthetic form of the plant-derived medicinal agent, plumbagin) and surgical castration caused regression of androgen dependent tumors in mice. These promising pre-clinical results led to this first-in-human study of PCUR-101 in combination with androgen deprivation therapy (ADT) in men with metastatic, castrate resistant PCa (mCRPC). Methods: The goal of this phase I multicenter trial was to determine the safety profile, maximum tolerated dose (MTD), recommended phase II dose, clinical activity, and pharmacokinetic (PK) parameters of PCUR-101. A 3 + 3 dose escalation design was employed. Patients (pts) in cohorts of 3 were treated with escalating doses of PCUR-101 (50 mg – 200 mg) orally once daily continuously. Cycles were 28 days. Exploratory correlates of IL-6 and urine polyamines were also included. Results: 12 pts (median age 75 [range 63-86]) with mCRPC on ADT were treated in the dose escalation cohorts. No DLTs were observed during treatment and the MTD was not reached. The most frequent adverse events (AEs) included diarrhea (11 pts; all grade 1 or 2), nausea (7 pts; all grade 1 or 2), vomiting (4 pts; all grade 1 or 2) and constipation (3 pts; all grade 1 or 2). No objective responses were observed but 1 pt had PSA decrease by > 50%. Pts remained on study treatment for a median of 10 weeks (range 3-32 weeks). 5 pts, with stable disease, remain on active treatment. PK data could not be fully evaluated due to issues with the PK assay. Analyses of IL-6 and putrescine levels in pt samples indicate that, as compared to no treatment, PCUR-101 treatment in each cycle was associated with decreases in their levels. Reasons for treatment discontinuation included disease progression (n = 4), adverse event (n = 1; nausea and vomiting), subject withdrawal (n = 1), and investigator or sponsor decision (n = 1). After treating 12 pts, the sponsor decided to stop the trial in order to reformulate the study drug to allow for higher dosing and to redevelop the PK assay. Conclusions: At the doses evaluated, PCUR-101 combined with ADT was seen to be safe and may prolong disease stability in men with mCRPC. A second phase I study is planned using a new drug formulation and PK assay. Clinical trial information: NCT03137758.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 7061-7061
Author(s):  
D. E. Tsai ◽  
S. Luger ◽  
A. Kemner ◽  
C. Andreadis ◽  
A. Loren ◽  
...  

7061 Background: In vitro, bexarotene inhibits the proliferation of non-M3 AML cell lines and induces differentiation of leukemic blasts. This phase I study was designed to evaluate the safety of escalating doses of bexarotene in patients with non-M3 AML and has completed enrollment. Methods: Bexarotene was administered daily until disease progression occurred. Dose escalation occurred in cohorts of 3–6 patients through 6 dose levels ranging from 100–400mg/m2. Results: 27 patients were enrolled: 19M/8F, median age 69 (range 51–82), 13 prior MDS, 12 primary refractory, median number of induction attempts 2, no prior chemotherapy 3, prior autologous stem cell transplant 5, 26 blood transfusion dependent, 18 platelet transfusion dependent, and 20 neutropenic. Despite prophylactic use of antihyperlipidemic agents, 4 patients developed grade ≥3 hypertriglyceridemia. Two patients developed a syndrome reminiscent of retinoic acid syndrome, consisting of dyspnea, pleural/pericardial effusions, and edema in the setting of a rising neutrophil count. This syndrome resolved with stopping bexarotene and initiating steroids. Evidence of activity was noted with bone marrow blasts decreasing to ≤5% in 4 patients. Seven patients showed evidence of neutrophil response (pretreatment median ANC 364/μL, range 28–1,242/μL, treatment ANC 3,540/μL, range 1,200–26,207/μL). Flow sorted peripheral blood neutrophils were collected from 3 of these patients and examined by FISH. Between 92–100% of neutrophils contained the patient's leukemic cytogenetic abnormality suggesting differentiation of the leukemic blasts. Eleven patients with platelet counts <100,000/μL had increases in their platelet counts >20,000/μL (peak range 40- 292x103/μL). Five of these patients with platelet counts <20,000/μL had improvement to 40–91,000/μL and became transfusion independent. Conclusions: Bexarotene is well tolerated at the dose levels studied. Evidence for clinical activity has been seen as exemplified by improvement in platelet counts, increased neutrophil counts and decreased bone marrow blasts. We postulate that bexarotene may induce leukemic blast differentiation in non-M3 AML and represent a novel non-cytotoxic treatment. No significant financial relationships to disclose.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 3501-3501 ◽  
Author(s):  
A. J. Wagner ◽  
D. H. Von Hoff ◽  
P. M. LoRusso ◽  
R. Tibes ◽  
K. E. Mazina ◽  
...  

3501 Background: The PI3K-PTEN-AKT signaling pathway is deregulated in a wide variety of cancers. GDC-0941 is a potent and selective oral inhibitor of the class I PI3K with 3 nM IC50 for the p110-alpha subunit in vitro and 28 nM IC50 in a cell-based pAKT assay and demonstrates broad activity in breast, ovarian, lung, and prostate cancer models. Methods: A Phase I dose escalation study using a 3+3 design was initiated in patients (pts) with solid tumors. GDC-0941 was given on d1, followed by 1 wk washout to study single-dose PK and PD markers. GDC-0941 was then administered qd on a 3 wk on, 1 wk off, schedule. Steady-state PK and PD were evaluated after 1 wk of continuous dosing. A separate concurrent dose-escalation arm with bid dosing was initiated after the third qd cohort. Results: Nineteen pts have been enrolled in 5 successive dose-escalation cohorts in the qd arm with dose levels up to 80 mg daily. Seven pts were enrolled in 2 cohorts in the bid arm at total daily doses of 60 and 80 mg. The most frequently reported drug-related AEs were Grade 1/2 nausea, fatigue, diarrhea, peripheral edema, and dysgeusia; no drug related grade >3 events have been reported. PK data suggest dose-proportional increases in Cmax and AUC. Potential signs of anti-tumor activity have been observed with a soft tissue sarcoma pt on-study for >176 days with stable disease (30 mg qd), an ovarian cancer pt with an on-study 2.8-fold decrease in CA-125 response to normal levels (30 mg bid) and a pt with endometrial cancer with a decrease in tumor FDG-PET uptake (80 mg qd). Conclusions: GDC-0941 is generally well-tolerated with potential signs of anti-tumor activity. Preliminary PK data suggest dose-proportional increases in exposure over the dose levels evaluated. Dose-escalation on both the qd and bid schedules continues with updated data to be presented. [Table: see text]


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3292-3292 ◽  
Author(s):  
Guillermo Garcia-Manero ◽  
Charles Chuah ◽  
George Wilding ◽  
Julie Chang ◽  
Srdan Verstovsek ◽  
...  

Abstract Abstract 3292 Background: SB939 is a novel orally bioavailable inhibitor of class 1, 2 and 4 histone deacetylases. In human tumor cell lines SB939 inhibits proliferation and promotes apoptosis at an IC50 of 0.1 – 1.3mM. Antitumor activity has been demonstrated in xenograft models of AML (MV4-11) and B-cell lymphoma (Ramos), as well as solid tumors. A phase I, open label, dose escalation study in patients with advanced hematologic malignancies was conducted to assess the safety, maximum tolerated dose (MTD), pharmacokinetics, pharmacodynamics and preliminary efficacy of SB939. Methods: SB939 was administered orally every other day 3 times a week for 3 consecutive weeks, in a 4-week cycle. Cohorts of patient were treated with escalating doses of SB939 starting from 10 mg. The MTD was defined as the lowest dose level with less than 2 DLTs. The recommended Phase 2 dose level was defined as one dose level below the MTD. PK and PD (Acetylated Histone 3 in PBMCs) samples were collected in the first cycle. Results: A total of 44 patients were enrolled. 23 patients during dose escalation at dose levels of 10 mg (n=1), 20 mg (n=1), 40 mg (n=6), 60 mg (n=3), 80 mg (n=3), 100 mg (n=3) and 120 mg (n=6). An additional 21 patients were enrolled as part of a cohort expansion at 100 mg. The median age was 70 yr (range 37–84 yr), 57% were male, 61% were caucasian and 27% asian. Median number of prior therapies was 2 (range 0–9), 16 % had a prior transplant. 89% had ECOG performance score of 0–1. The median number of doses received was 17. DLTs included prolonged QTc at 40 mg and neutropenic sepsis at 120 mg. The MTD as defined was not reached; 120 mg was declared as MTD due to the requirement for dose reduction after multiple cycles of treatment. 100 mg was determined to be the recommended Phase II dose. 24 patients, MDS (n=11), AML (n=12), and lymphoma (n=1) were treated at the 100 mg dose level. SB939 was generally well tolerated. Grade 1–2 events included nausea (45%), fatigue (44%), diarrhea (36%), anorexia (34%) and vomiting (30%). Grade 3–4 adverse events included thrombocytopenia (39%), anemia (23%), pneumonia (23%), febrile neutropenia (20%), fatigue (16%), hypokalemia (11%), and neutropenic sepsis (11%). Samples for pharmacokinetics were drawn prior to dosing and 0.5, 1, 1.5, 2, 3, 4, 6, 8, 24 ± 2 and 30 ± 2 hours after dosing on days 1 and 15 of Cycle 1. Levels of SB939 in plasma were determined using a validated LC-MS/MS method and Non-Compartmental Analysis used WinNonlin, version 5.2 (Pharsight). SB939 was rapidly absorbed with mean Tmax ranging between 0.5–1.3 h; the mean elimination half-life ranged between 6–17 hrs. The Cmax and AUC (0-∞) increased dose-proportionally in the range of doses tested. There was no accumulation of SB939 on day 15 following repeated dosing. Concentrations above IC50 of SB939 for HDAC 1, 2, and 4 were reached at all doses and increased acetylation of H3 was observed in PBMCs across all dose levels. 1 PR (80 mg) and 1 CR (120 mg) were observed in 2 patients with AML with durations of 362 and 206 days respectively. Stable disease for more than 2 cycles was seen in 7 patients, 3 with IPSS intermediate or high risk MDS (duration 72–134 d) and 4 with AML (duration 56–354 d). Conclusions: SB939 demonstrated excellent PK properties and target inhibition and was generally very well tolerated. Toxicities were mild to moderate and similar to some but not all toxicities seen with other HDAC inhibitors. The MTD as defined for this regimen of SB939 in patients with hematologic malignancies was not reached and 100mg is the recommended dose, indicating a favorable therapeutic index. Response data particularly in higher risk MDS and AML encourage further exploration of the therapeutic benefit of SB939 in combination with other anti-cancer therapies. Disclosures: Ethirajulu: S*BIO: Employment. Zhu:S*BIO: Employment.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4048-4048 ◽  
Author(s):  
Jesus G. Berdeja ◽  
Lowell Hart ◽  
Ruth Lamar ◽  
Patrick Murphy ◽  
Susan Morgan ◽  
...  

Abstract Abstract 4048 Background: Proteasome inhibitors (PI) such as bortezomib and carfilzomib (CFZ) have improved the treatment of MM; however, resistance invariably develops and MM remains an incurable disease. Changes in histone modification are commonly found in human cancers including MM, and several histone deacetylase inhibitors (HDACi) are in clinical development. Preclinical studies demonstrate synergistic anti-MM activity of the combination of HDACi and PI through dual inhibition of the proteasome and aggresome pathways. Panobinostat (PAN) is an oral pan-HDACi which has shown synergy with bortezomib in clinical studies. This Phase I/II trial will determine the maximum tolerated dose (MTD) and evaluate the safety and efficacy of the combination of PAN and CFZ in relapsed/refractory pts with MM. When the MTD has been determined, the trial will continue to enroll pts with relapsed/refractory MM at the recommended dose. Methods: The dose escalation portion of the study will determine the maximum tolerated dose (MTD) of 4 planned dose levels with the combination of CFZ and PAN using a standard 3+3 dose escalation design. Dose modifications are not permitted during cycle 1 unless a pt experiences a dose limiting toxicity (DLT). DLTs are defined as: G4 neutropenia, febrile neutropenia, G4 thrombocytopenia or G3 thrombocytopenia with bleeding, G2 neuropathy with uncontrolled pain, any G3 non-hematologic drug-related toxicity requiring a dose reduction, or pts who are unable to receive 75% of the required dose due to toxicity. PAN is administered orally three times weekly during weeks 1 and 3 of each cycle (Days 1, 3, 5, 15, 17, 19). CFZ is administered IV over 10 min for doses < 27mg/m2 and over 30 min for doses >27 mg/m2 on Days 1, 2, 8, 9, 15, and 16 of each 28-day cycle. AEs are assessed according to CTCAE Version 4 and responses are assessed using IMWG criteria (plus MRs as per the EBMT criteria). Results: Ten pts have been enrolled to date across 3 dose levels and completed the DLT window. The dose levels explored to date are: level 1, CFZ 20/27 and PAN 20 mg; level 2 CFZ 20/36 and PAN 20 mg; and level 3, CFZ 20/45 and PAN 20 mg. The median age of pts was 65.5 (range 42–75). The median number of prior therapies was 3 (range 1–7); all pts had prior bortezomib (10% bortezomib refractory) and 40% were refractory to their last treatment. Nine pts are currently on study and 1 pt from DL1 discontinued due to progressive disease. No DLTs have been observed to date, there have been no dose reductions and there have been no deaths on study. Treatment related hematologic AEs were G3 thrombocytopenia observed in 40% of pts in dose levels 1–3, followed by neutropenia in 10% of pts from DL 1. No G4 hematologic and only 1 G3 non-hematologic AEs were observed. The most common treatment related non-heme AEs were GI with grade 1/2 nausea and vomiting occurring in 70% of pts (60% G1 & 10% G2), diarrhea in 60% of pts (40% G1, 10% G2 & 10% G3). Other mild to moderate adverse events occurring in >1 pt were; fatigue 50% of pts (30% G1, 20% G2), fever/chills 50% (G1), dry mouth 30% (G1), congestion 20% (G2), dysgusia 20% (10% G1, 10% G2), epitaxis 20% (G2), hypocalcemia 20% (G1), and liver function 20% (G1). Grade 2 peripheral neuropathy was reported in 1 patient. Of the 10 pts treated, 9 are evaluable for response (10% VGPR, 40% PR, 10% MR, and 30% SD). The median number of cycles for the evaluable pts is 5 (range 2–8). Conclusions: The combination of PAN and CFZ is well tolerated in relapsed/refractory MM patients. There have been no DLTs or dose reductions reported to date. MTD has not been reached at the current dose of CFZ 20/45 and PAN 20. The majority of AEs reported were manageable and expected. Early efficacy results are encouraging, and dose escalation to CFZ 20/45 and PAN 30 is planned. Disclosures: Off Label Use: Off-label use of the combo Panobinostat and Carfilomib in the treatment of relapsed/refractory Multiple Myeloma.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 7142-7142 ◽  
Author(s):  
C. S. Rocha Lima ◽  
S. V. Orlov ◽  
J. Garst ◽  
G. M. Manikhas ◽  
A. Dowlati ◽  
...  

7142 Background: Talotrexin is a nonpolyglutamatable antifolate which has demonstrated improved antitumor activity in a broad spectrum of cancer models by targeting DHFR to inhibit tumor growth. Talotrexin binds more tightly (15-fold, Ki 0.35 pM) to DHFR than methotrexate (MTX). In lung cancer cell lines, talotrexin inhibits tumor cell proliferation at sub- to low nanomolar concentrations and is more potent than MTX in all cell lines tested. We conducted a dose escalation study of talotrexin administered as a 5-minute infusion on Days 1, 8, on a 21-day cycle. The primary objectives of this study are to determine the maximum tolerated dose (MTD), pharmacokinetic (PK) profile, as well as the safety and efficacy. Methods: Patients with confirmed NSCLC stage III/IV or recurrent/refractory disease, failed two-lines of therapy ± an EGFR TK inhibitor, ECOG PS 0–2, were enrolled. The starting dose was based on a Phase I solid tumor trial. Three patients were treated at each dose level in the absence of a dose limiting toxicity (DLT). Intrapatient dose escalation was allowed. All patients received folic acid and vitamin B12. Safety assessments were performed by standard laboratory and clinical DLT definitions. Responses were assessed based on RECIST criteria. Plasma talotrexin concentrations were analyzed by a LC/MS/MS method. Concentration-time profiles of talotrexin were analyzed by noncompartmental methods using WinNonlin. Results: Eighteen heavily pre-treated patients were enrolled and received a total of 61 cycles (median 3.4, range 1–7) of talotrexin at 5 dose levels (27–270 mg/m2/cycle), Male/Female: 14/4. The tentative MTD is 90 mg/m2/dose, with mucositis being the predominate DLT. Two partial responses and 9 stable diseases were observed. Talotrexin exhibits linear PK profile across the 5 dose levels evaluated. PK data (16 patients), Cmax and AUC0-∞ exhibited linear PK. Overall mean values (± SD) were CL, 1.31 ± 0.31 L/hr/m2, T1/2z, 6.5 ± 1.4 h, Vss 8.9 ± 2.5 L/m2. Conclusions: Talotrexin demonstrated acceptable tolerability with encouraging activity in relapsed or refractory NSCLC patients over multiple cycles of therapy. Phase II studies in NSCLC are planned. [Table: see text]


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 8006-8006 ◽  
Author(s):  
Sagar Lonial ◽  
Niels W.C.J. van de Donk ◽  
Rakesh Popat ◽  
Jeffrey A. Zonder ◽  
Monique C. Minnema ◽  
...  

8006 Background: IBER is a novel cereblon E3 ligase modulator (CELMoD) with enhanced tumoricidal and immunostimulatory activities. Preclinically, IBER overcomes immunomodulatory drug (IMiD) resistance and has synergy with daratumumab (DARA), bortezomib (BORT), and DEX. Methods: A phase 1b/2a multicenter, open-label, dose-escalation study evaluated the maximum tolerated dose (MTD), recommended phase 2 dose (RP2D), safety, and preliminary efficacy of IBER. Eligible pts had RRMM and must have received ≥ 2 prior regimens including lenalidomide (LEN) and/or pomalidomide (POM), and a proteasome inhibitor (PI). All pts had progressed on or within 60 days of last MM therapy. Escalating doses of IBER were given on days 1–21, in combination with DEX 40 mg (20 mg in pts age > 75 years) on days 1, 8, 15, and 22, of each 28-day cycle. Dose escalation was reviewed by a dose escalation committee. Results: As of Jan 2019,58 pts received IBER + DEX. Median age was 64.5 years (range 33–79), and median number of prior regimens was 5 (2–12). Prior therapies included autologous stem cell transplant (79%), LEN (100%), POM (69%), PIs (100%), and DARA (66%). IBER dose ranged from 0.3 to 1.2 mg; MTD/RP2D was not reached. Median duration of therapy was 12+ weeks (range 4–109). Grade 3–4 adverse events (AEs) were reported in 41 (72%) pts and were not related to dose. Grade 3–4 neutropenia, thrombocytopenia, neuropathy, and fatigue occurred in 26%, 11%, 2%, and 0% pts, respectively. Three pts discontinued treatment due to AEs. Clinical activity occurred early and was observed across all dose levels (Table); 20 of 51 pts remain on treatment (2–27+ cycles). Conclusions: IBER + DEX showed favorable efficacy and safety in heavily pretreated pts with RRMM who failed multiple prior therapies. This study is ongoing, including combinations of IBER with DARA or BORT. Clinical trial information: NCT02773030. [Table: see text]


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 6621-6621
Author(s):  
Philip C. Amrein ◽  
Eyal C. Attar ◽  
Amir Tahmasb Fathi ◽  
Steven L McAfee ◽  
Martha Wadleigh ◽  
...  

6621 Background: Both bortezomib (Bz) and lenalidomide have clinical activity in patients with MDS and AML. We conducted a phase I dose escalation study to determine the maximal tolerated dose (MTD) of Bz in combination with lenalidomide. Methods: Patients with MDS (IPSS score ≥ 0.5 or therapy-related) received Bz by IV bolus on Days 1, 4, 8, and 11 and lenalidomide 10 mg/day PO on Days 1-21 in 28 day cycles for up to 9 cycles. Three doses of Bz were tested (0.7, 1.0, or 1.3 mg/m2). Cohorts consisted of 3-6 patients; the dose of Bz was escalated if there were < 2 dose limiting toxicities (DLTs). Growth factor support and transfusions were permitted. Dose limiting toxicities (DLTs) were assessed during the first cycle and were defined as severe neutropenia (absolute neutrophil count ≤ 250/ul), thrombocytopenia (platelet count < 10,000/ul), grade ≥ 2 neurotoxicity, or other grade ≥ 3 non-hematologic toxicity. Following determination of the MTD, enrollment opened to patients with relapsed and refractory AML and those with untreated high risk disease for whom induction therapy was not indicated. Responses were assessed by IWG 2006 criteria for MDS and IWG 2003 criteria for AML. Results: 23 patients (14 men) were enrolled; one patient was inevaluable due to disease progression prior to starting protocol therapy. The median age was 73 years (range 54-87). There was 1 DLT observed, neutropenia, in 6 patients treated with 1.0 mg/m2 Bz and no DLTs at 0.7 or 1.3 mg/m2. The median number of cycles was 2 (range 2-9). Grade ≥ 3 toxicities possibly attributable to the treatment at any dose level were: anemia (2), thrombocytopenia (10), leukopenia (3), infection (1), rash (2), dyspnea (1), dizziness (1), hypotension (1), pneumonia (2) and neuropathy (1). Among the 14 patients with MDS, 1 patient with RARS experienced a CR and 2 with RAEB-2 experienced marrow CR (mCR). Among the 8 patients with AML, there was 1 CR. Conclusions: The maximal tested dose of Bz (1.3 mg/m2) in combination with lenalidomide is safe. Responses were seen in MDS and high risk AML. Future testing of this regimen is planned.


Sign in / Sign up

Export Citation Format

Share Document